Language selection

Search

Patent 2697363 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2697363
(54) English Title: MODULATION OF SYNAPTOGENESIS
(54) French Title: MODULATION DE LA SYNAPTOGENESE
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 39/395 (2006.01)
  • A61K 38/17 (2006.01)
  • A61K 45/00 (2006.01)
  • G01N 33/53 (2006.01)
  • G01N 33/68 (2006.01)
  • A61K 31/713 (2006.01)
(72) Inventors :
  • EROGLU, CAGLA (United States of America)
  • BARRES, BEN A. (United States of America)
(73) Owners :
  • THE BOARD OF TRUSTEES OF THE LELAND STANFORD JUNIOR UNIVERSITY (United States of America)
(71) Applicants :
  • THE BOARD OF TRUSTEES OF THE LELAND STANFORD JUNIOR UNIVERSITY (United States of America)
(74) Agent: BORDEN LADNER GERVAIS LLP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2008-08-14
(87) Open to Public Inspection: 2009-03-05
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2008/009747
(87) International Publication Number: WO2009/029173
(85) National Entry: 2010-02-22

(30) Application Priority Data:
Application No. Country/Territory Date
60/966,073 United States of America 2007-08-23
61/052,551 United States of America 2008-05-12

Abstracts

English Abstract





The present invention describes methods and compositions for modulating
synaptogenesis and axon and/or dendritic
growth. The methods include the use of agents that modulate a thrombospondin
and/or an a26 subunit of a calcium channel.


French Abstract

L'invention concerne des procédés et des compositions de modulation de la synaptogenèse et de la croissance axonique et/ou dendritique. Ces procédés consistent à utiliser des agents qui modulent une thrombospondine et/ou une sous-unité a2d de canal calcique.

Claims

Note: Claims are shown in the official language in which they were submitted.





WHAT IS CLAIMED IS:



1. A method of promoting synaptogenesis in an individual comprising
administering
to the individual in need of synaptogenesis an effective dose of a polypeptide
comprising at least
one thrombospondin EGF-like domain, wherein the polypeptide is not a
thrombospondin, and
wherein synapse formation in the individual is increased.


2. The method of Claim 1, wherein the thrombospondin EGF-like domain is a
polypeptide derived from a thrombospondin isotype of from about 35 to about 65
amino acids in
length, comprising at least 6 cysteine amino acids, where the main structure
is a two-stranded
beta-sheet followed by a loop to a C-terminal short two-stranded sheet.


3. The method of Claim 2, wherein the thrombospondin EGF-like domain has at
least 95% sequence identity to human TSP1, amino acids 551-586, 588-636, or
650-689, human
TSP2 amino acids 553-588, 590-635, or 652-691, human TSP3 amino acids 316-368,
370-412,
or 418-455, human TSP4 amino acids 290-324, 326-377, 379-418 or 424-461, or
human
cartilage oligomeric matrix amino acids 87-126, 127-179, 180-222, or 225-267.


4. The method of Claim 3, wherein the polypeptide comprises at least two
thrombospondin EGF-like domains.


5. The method of Claim 4, wherein the polypeptide comprises at least three
thrombospondin EGF-like domains.


6. The method of Claim 5, wherein the polypeptide lacks thrombospondin
sequences
other than the EGF-like domains.


7. The method of Claim 1, wherein the individual has suffered synapse loss as
a
result of senescence.


8. The method of Claim 1, wherein the individual has suffered synapse loss as
a



88




result of Alzheimer's disease, Parkinson's disease, ALS, multiple sclerosis,
or glaucoma.

9. The method of Claim 1, wherein the individual has suffered a macular
degeneration, a hearing loss, a diabetic neuropathy, or a chemotherapy induced
neuropathy.

10. The method of Claim 1, wherein the individual has suffered synapse loss as
a
result of a psychiatric disorder selected from the group consisting of
depression, schizophrenia,
autism, and aggression.


11. The method according to Claim 1, wherein said synapse formation is at a
neuromuscular junction.


12. A method of treating or preventing pain in an individual comprising
administering
to an individual an effective amount of a thrombospondin antagonist agent
which inhibits
synaptogenesis activity of a thrombospondin.


13. The method of Claim 12, wherein the agent binds to a thrombospondin and
blocks
the interaction between the thrombospondin and one or more calcium channel
subunits selected
from the group consisting of .alpha.2.delta.1,
.alpha.2.delta.2,.alpha.2.delta.3, and .alpha.2.delta.4


14. The method of Claim 13, wherein the agent is an antibody that specifically
binds
to the thrombospondin.


15. The method of Claim 14, wherein the agent is an antibody that specifically
binds
to an EGF-like domain of the thrombospondin.


16. The method of Claim 14, wherein the agent is an antibody that specifically
binds
to the third EGF-like domain of the thrombospondin.


17. The method of Claim 14, wherein the thrombospondin is TSP1, TSP2, TSP3,
TSP4, or cartilage oligomeric matrix.



89




18. The method of Claim 13, wherein the agent is a scaffold-derived binding
protein
that specifically binds to the thrombospondin.


19. The method of Claim 13, wherein the agent is a siRNA, an antisense RNA, or
a
microRNA that specifically inhibits expression of the thrombospondin.


20. The method of Claim 19, wherein the expression of TSP1, TSP2, TSP4, or
cartilage oligomeric matrix is inhibited.


21. The method of Claim 13, wherein the agent is a polypeptide comprising an
extracellular portion of a calcium channel subunit .alpha.2.delta.1.


22. The method of Claim 21, wherein the polypeptide comprises the amino acids
of
about 253 to about 430 of human .alpha.2.delta.l (VWFA domain).


23. The method of Claim 13, wherein the agent is a polypeptide comprising an
extracellular portion of a calcium channel subunit .alpha.2.delta.2.


24. The method of Claim 23, wherein the polypeptide comprises the amino acids
of
about 291 to about 469 of human .alpha.2.delta.2 (VWFA domain).


25. The method of Claim 13, wherein the agent is a polypeptide comprising an
extracellular portion of a calcium channel subunit .alpha.2.delta.3.


26. The method of Claim 25, wherein the polypeptide comprises the amino acids
of
about 256 to about 438 of human .alpha.2.delta.3 (VWFA domain).


27. The method of Claim 13, wherein the agent is a polypeptide comprising an
extracellular portion of the calcium channel subunit .alpha.2.delta.4.



90




28. The method of Claim 27, wherein the polypeptide comprises the amino acids
of
about 291 to about 473 of human .alpha.2.delta.4 (VWFA domain).


29. The method of any one of Claims 21, 23, 25, and 27, wherein the
polypeptide is
an immunoadhesin.


30. The method of claim 12, wherein the pain is somatic pain, neuropathic
pain,
visceral pain, cancer pain, breakthrough cancer pain, inflammatory pain, post
operative pain,
bone pain, joint pain, migraine pain, or phantom pain.


31. The method of claim 12, wherein the pain is allodynia or hyperalgesia.


32. A method of treating or preventing pain in an individual comprising
administering
to an individual an effective amount of an antibody that specifically binds to
a VWFA domain of
a calcium channel subunit selected from the group consisting of
.alpha.2.delta.1, .alpha.2.delta.2, .alpha..alpha.2.delta.3, and
.alpha.2.delta.4,
and blocks the interaction between a thrombospondin and said calcium channel
subunit.


33. The method of Claim 32, wherein the pain is somatic pain, neuropathic
pain,
visceral pain, cancer pain, breakthrough cancer pain, inflammatory pain, post
operative pain,
bone pain, joint pain, migraine pain, or phantom pain.


34. A method of treating epilepsy in an individual comprising administering to
the
individual an effective amount of an antibody that specifically binds to a
VWFA domain of a
calcium channel subunit selected from the group consisting of
.alpha.2.delta.1, .alpha.2.delta.2, .alpha.2.delta.3, and .alpha.2.delta.4,
and
blocks the interaction between a thrombospondin and said calcium channel
subunit.


35. A method of promoting axonal growth in an individual comprising
administering
to an individual in need thereof an effective amount of a thrombospondin
antagonist.


36. The method of claim 35, wherein the individual has suffered a spinal cord
injury.


91




37. The method of Claim 35, wherein the individual has suffered axonal or
dendritic
degeneration as a result of Alzheimer's disease, Parkinson's disease, ALS, or
multiple sclerosis.

38. The method of Claim 35, wherein the individual has suffered a macular
degeneration, a hearing loss, a diabetic neuropathy, or a chemotherapy induced
neuropathy.


39. The method of Claim 35, wherein the individual has suffered axonal or
dendritic
degeneration as a result of a psychiatric disorder selected from the group
consisting of
depression, schizophrenia, autism, and aggression.


40. A method for treating a disorder characterized by excess of calcium influx
in an
individual, comprising administering to the individual an effective amount of
an agent that
specifically binds to a thrombospondin and blocks the interaction between the
thrombospondin
and a calcium subunit selected from the group consisting of .alpha.2.delta.1,
.alpha.2.delta.2, .alpha.2.delta.3, and .alpha.2.delta.4.


41. The method of Claim 40, wherein the disorder is selected from the group
consisting of muscle spasm, migraine, stroke, and Parkinson's disease.


42. A method of screening a candidate agent for activity in enhancing
synaptogenesis,
the method comprising:
a) measuring binding of a candidate agent to an .alpha.2.delta. polypeptide or
a thrombospondin
EGF-like domain;
b) quantitating formation of synapses in a neural cell culture in the presence
of the
candidate agent if the candidate agent binds to the .alpha.2.delta.
polypeptide or the thrombospondin EGF-
like domain in step a), wherein an increased formation of synapses in the
presence the candidate
agent as compared to the formation of synapses in the absence of the candidate
agent indicates
that the candidate agent has the activity in enhancing synaptogenesis.


43. A method of screening a candidate agent for activity in inhibiting
synaptogenesis,
the method comprising:
a) measuring binding of a candidate agent to an .alpha.2.delta. polypeptide or
a thrombospondin


92




EGF-like domain;
b) quantitating formation of synapses in a neural cell culture in the presence
of the
candidate agent and a thrombospondin agonist if the candidate agent binds to
the .alpha.2.delta.
polypeptide or the thrombospondin EGF-like domain in step a), wherein a
decreased formation
of synapses in the presence the candidate agent as compared formation of
synapses in the
absence of the candidate agent indicates that the candidate agent has the
activity in inhibiting
synaptogenesis.



93

Description

Note: Descriptions are shown in the official language in which they were submitted.



CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
MODULATION OF SYNAPTOGENESIS

RELATED APPLICATIONS
[0001] This application claims priority benefit of U.S. Provisional
Application Nos.
60/966,073, filed August 23, 2007 and 61/052,55 1, filed May 12, 2008, which
are hereby
incorporated by reference in their entirety.

TECHNICAL FIELD
[0002] The present invention relates generally to compositions and methods for
modulating
synaptogenesis and axon and dendritic growth. Specifically, the invention
includes use of agents
that modulate interactions between a thrombospondin and an alpha 2 delta
subunit of a calcium
channel.

BACKGROUND OF THE INVENTION
[0003] Synapses are specialized cell adhesions that are the fundamental
functional units of
the nervous system, and they are generated during development with amazing
precision and
fidelity. During synaptogenesis, synapses form, mature, and stabilize and are
also eliminated by
a process that requires intimate communication between pre- and postsynaptic
partners. In
addition, there may be environmental determinants that help to control the
timing, location, and
number of synapses.
[0004] Synapses occur between neuron and neuron and, in the periphery, between
neuron
and effector cell, e.g. muscle. Functional contact between two neurons may
occur between axon
and cell body, axon and dendrite, cell body and cell body, or dendrite and
dendrite. It is this
functional contact that allows neurotransmission. Many neurologic and
psychiatric diseases are
caused by pathologic overactivity or underactivity of neurotransmission; and
many drugs can
modify neurotransmission, for examples hallucinogens, antipsychotics, anti-
schizophrenia,
tranquilizers, sedatives, anesthetics, pain drugs, Alzheimer's disease drugs,
and Parkinson's
disease drugs.
[0005] During recent years, a great deal of effort has been made by
investigators to
characterize the function of synaptic proteins, which include synaptotagmin,
syntexin,
synaptophysin, synaptobrevin, and the synapsins. These proteins are involved
in specific aspects
of synaptic function, e.g. synaptic vesicle recycling or docking, and in the
organization of

1


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
axonogenesis, differentiation of presynaptic terminals, and in the formation
and maintenance of
synaptic connections. See, for example, U.S. Patent Application Publication
No. 2006/0019880.
[0006] Only by establishing synaptic connections can nerve cells organize into
networks
and acquire information processing capability such as learning and memory.
Synapses are
progressively reduced in number during normal aging, and are severely
disrupted during
neurodegenerative diseases. Therefore, finding molecules capable of creating
and/or maintaining
synaptic connections is an important step in the treatment of
neurodegenerative diseases.
Conversely, excessive synapse formation may be associated with stroke and
psychiatric
disorders. The modulation of synapse formation is of great interest for the
treatment of a variety
of nervous system disorders.
[0007] US 2006/0019880 discloses that thrombospondins can trigger synapse
formation and
a thrombospondin agonist or an antagonist can be used to promote or inhibit
synaptogenesis in a
patient in need of synaptogenesis promotion or inhibition.
[0008] All references, publications, and patent applications disclosed herein
are hereby
incorporated by reference in their entirety.

SUMMARY OF THE INVENTION
[0009] The present invention provides methods for promoting synaptogenesis in
an
individual comprising administering to the individual in need of
synaptogenesis an effective dose
of a polypeptide comprising at least one thrombospondin EGF-like domain,
wherein the
polypeptide is not a thrombospondin, and wherein synapse formation in the
individual is
increased. In some embodiments, the polypeptide binds and/or activates a
calcium channel
subunit selected from the group consisting of a26 1, a262, a263, and a284.
[0010] In some embodiments, the thrombospondin EGF-like domain is a
polypeptide
derived from a thrombospondin isotype of from about 35 to about 65 amino acids
in length,
comprising at least 6 cysteine amino acids, where the main structure is a two-
stranded beta-sheet
followed by a loop to a C-terminal short two-stranded sheet. In some
embodiments, the
thrombospondin EGF-like domain has at least 95% sequence identity to human
TSP1, amino
acids 551-586, 588-636, or 650-689, human TSP2 amino acids 553-588, 590-635,
or 652-691,
human TSP3 amino acids 316-368, 370-412, or 418-455, human TSP4 amino acids
290-324,
326-377, 379-418 or 424-461, or human cartilage oligomeric matrix amino acids
87-126, 127-
179, 180-222, or 225-267. In some embodiments, the polypeptide comprises at
least two

2


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
thrombospondin EGF-like domains. In some embodiments, the polypeptide
comprises at least
three thrombospondin EGF-like domains. In some embodiments, the polypeptide
lacks
thrombospondin sequences other than the EGF-like domains. In some embodiments,
the
individual has suffered synapse loss as a result of senescence. In some
embodiments, the
individual has suffered synapse loss as a result of Alzheimer's disease,
Parkinson's disease,
ALS, multiple sclerosis, spinal cord injury, enteric nervous system disorders
or glaucoma. In
some embodiments, the individual has suffered a macular degeneration, a
hearing loss, a diabetic
neuropathy, or a chemotherapy induced neuropathy. In some embodiments, the
individual has
suffered synapse loss as a result of a psychiatric disorder selected from the
group consisting of
depression, schizophrenia, autism, and aggression. In some embodiments,
synapse formation is
at a neuromuscular junction. In some embodiments, synapse formation is at a
muscle.
[0011] The invention also provides methods for treating a disorder caused by
the presence
of excess, hyperactive, abnormally connected or dysfunctional synapses
comprising
administering to an individual an effective amount of a thrombospondin
antagonist agent which
inhibits synaptogenesis activity of a thrombospondin. In some embodiments, the
agent binds to a
thrombospondin and blocks the interaction between the thrombospondin and one
or more
calcium channel subunits selected from the group consisting of a26 l, a282,
a283, and a284.
[0012] The invention also provides methods for treating or preventing pain in
an individual
comprising administering to the individual an effective amount of a
thrombospondin antagonist
agent which inhibits an activity of a thrombospondin.
[0013] In some embodiments, the thrombospondin antagonist agent binds to a
thrombospondin and blocks the interaction between the thrombospondin and one
or more
calcium channel subunits selected from the group consisting of a28I, a262,
a263, and a284. In
some embodiments, the agent is an antibody that specifically binds to the
thrombospondin. In
some embodiments, the agent is an antibody that specifically binds to an EGF-
like domain of the
thrombospondin. In some embodiments, the agent is an antibody that
specifically binds to the
third EGF-like domain of the thrombospondin. In some embodiments, the
thrombospondin is
TSP1, TSP2, TSP3, TSP4, or cartilage oligomeric matrix. In some embodiments,
the antibody
specifically binds to TSP l, TSP2, TSP3, TSP4, or cartilage oligomeric matrix.
In some
embodiments, the antibody specifically binds to more than one members of
thrombospondin. In

3


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
some embodiments, the agent is a protein scaffold for an antibody mimic or a
scaffold-derived
binding protein.
[0014] In some embodiments, the agent is a polypeptide comprises an
extracellular portion
of a calcium channel subunit selected from the group consisting of a28 l,
a282, a283, and a254,
wherein polypeptide binds to a thrombospondin and blocks the interaction
between the
thrombospondin and the calcium channel subunit. In some embodiments, the agent
is a
polypeptide comprising the a2 portion of the calcium channel subunit a261. In
some
embodiments, the polypeptide comprises the amino acids of about 253 to about
430 of human
a281 (VWFA domain). In some embodiments, the agent is a polypeptide comprising
the a2
portion of the calcium channel subunit human a262. In some embodiments, the
polypeptide
comprises the amino acids of about 291 to about 469 of human a282 (VWFA
domain). In some
embodiments, the agent is a polypeptide comprising the a2 portion of the
calcium channel
subunit a283. In some embodiments, the polypeptide comprises the amino acids
of about 256 to
about 438 of human a263 (VWFA domain). In some embodiments, the agent is a
polypeptide
comprising the a2 portion of the calcium channel subunit a284. In some
embodiments, the
polypeptide comprises the amino acids of about 291 to about 472 of human a254.
In some
embodiments, the polypeptide comprises the amino acids of about 291 to about
473 of human
a264 (VWFA domain). In some embodiments of any of the method claims, the
polypeptide is an
immunoadhesin.
[0015] In some embodiments, the agent is an siRNA, an antisense RNA, or a
microRNA
that specifically inhibits expression of the thrombospondin. In some
embodiments, the
expression of TSP1, TSP2, TSP4, or cartilage oligomeric matrix is inhibited.
[0016] The invention further provides methods for treating or preventing pain
in an
individual comprising administering to an individual an effective amount of an
antibody that
specifically binds to a VWFA domain of a calcium channel subunit selected from
the group
consisting of a28 1, a262, a263, and a254, and blocks the interaction between
a thrombospondin
and said calcium channel subunit. In some embodiments, the antibody
specifically binds to a
VWFA domain of the calcium channel subunit selected from the group consisting
of a28 1, a282,
a283, and a284.
[0017] In some embodiments, the antibody binds to a region comprising from
about amino
acids 253 to about 430 of human a281 (VWFA domain). In some embodiments, the
antibody

4


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
binds to a region comprising from about amino acids 291 to about 469 of human
a262 (VWFA
domain). In some embodiments, the antibody binds to a region comprising from
about amino
acids 256 to about 438 of human a253 (VWFA domain). In some embodiments, the
antibody
binds to a region comprising from about amino acids 291 to about 473 of human
a284 (VWFA
domain).
[0018] Different type of pains can be treated by the methods described herein.
The types of
pain include, but are not limited to, somatic pain; pain associated with
ligaments, tendons, bones,
blood vessels, fasciae, or muscles; visceral pain; any pain caused by injury
to the nervous
system, chemotherapy, radiation, surgery, tumor compressing, or accident;
cancer pain;
inflammatory pain; post operative pain; migraine pain; phantom pain;
allodynia; or hyperalgesia.
In some embodiments, the pain can be either acute or chronic.
[0019] The invention also provides methods for treating epilepsy in an
individual
comprising administering to the individual an effective amount of an antibody
that specifically
binds to a VWFA domain of a calcium channel subunit selected from the group
consisting of
a28 1, a282, a283, and a264, and blocks the interaction between a
thrombospondin and said
calcium channel subunit. In some embodiments, the antibody specifically binds
to a VWFA
domain of the calcium channel subunit selected from the group consisting of
a26 1, a262, a283,
and a2S4.
[0020] The invention provides methods for promoting axonal growth in an
individual
comprising administering to an individual in need thereof an effective amount
of a
thrombospondin antagonist agent. In some embodiments, the individual has
suffered a spinal
cord injury. In some embodiments, the individual has suffered axonal or
dendritic degeneration
as a result of Alzheimer's disease, Parkinson's disease, ALS, or multiple
sclerosis. In some
embodiments, the individual has suffered a macular degeneration, a hearing
loss, a diabetic
neuropathy, cancer-induced neuropathy, radiation-induced neuropathy, or a
chemotherapy-
induced neuropathy. In some embodiments, the individual has suffered axonal or
dendritic
degeneration as a result of a psychiatric disorder selected from the group
consisting of
depression, schizophrenia, autism, anxiety, and aggression. Any thrombospondin
agent
described herein may be administered.
[0021] The invention also provides methods for treating a disorder
characterized by excess
of calcium influx in an individual, comprising administering to the individual
an effective



CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
amount of an agent that blocks the interaction between a thrombospondin and a
calcium subunit
selected from the group consisting of a281, a252, a283, and a264. In some
embodiments, the
disorder is selected from the group consisting of muscle spasm, migraine,
stroke, Alzheimer's
disease, and Parkinson's disease. In some embodiments, the agent specifically
binds to the
thrombospondin.
[0022] The invention further provides methods for screening a candidate agent
for activity
in enhancing synaptogenesis, the method comprising: a) measuring binding of a
candidate agent
to an a281, a282, a283, or a264 polypeptide or a thrombospondin EGF-like
domain; b)
quantitating formation of synapses in a neural cell culture in the presence of
the candidate agent
if the candidate agent binds to the a28 polypeptide or the thrombospondin EGF-
like domain in
step a), wherein an increased formation of synapses in the presence the
candidate agent as
compared to the formation of synapses in the absence of the candidate agent
indicates that the
candidate agent has the activity in enhancing synaptogenesis.
[00231 The invention also provides methods for screening a candidate agent for
activity in
inhibiting synaptogenesis, the method comprising: a) measuring binding of a
candidate agent to
an a25 1, a262, a283, or a284 polypeptide or a thrombospondin EGF-like domain;
b) quantitating
formation of synapses in a neural cell culture in the presence of the
candidate agent and a
thrombospondin agonist if the candidate agent binds to the a26 polypeptide or
the
thrombospondin EGF-like domain in step a), wherein a decreased formation of
synapses in the
presence the candidate agent as compared formation of synapses in the absence
of the candidate
agent indicates that the candidate agent has the activity in inhibiting
synaptogenesis.
[0024] It is to be understood that one, some, or all of the properties of the
various
embodiments described herein may be combined to form other embodiments of the
present
invention.

BRIEF DESCRIPTION OF THE DRAWINGS
[0025] Figure IA: Pentameric Subgroup B Thrombospondins (TSPs) are
Synaptogenic.
TSPs are divided into two subgroups according to their domain structure and
oligomerization
state. Subgroup A TSPs (TSPs 1-2) are trimeric. Subgroup B TSPs (TSPs 3-5) are
pentameric.
The domain structure of subgroup B TSPs differ at the N-terminal part of the
molecule. They
possess a different N-terminal domain (black oval), and lack the procollagen
(small square) and
properdin-like repeats (rectangles), which are present in Subgroup A TSPs.
Whereas all TSPs

6


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
share a common domain structure at the C-terminal end composed of three EGF-
like repeats
(small ovals), thirteen calcium binding repeats (long rectangle), and a C-
terminal L-lectin like
globular domain (light-colored square).
[0026] Figures 1B-F: Pentameric Subgroup B Thrombospondins (TSPs) are
Synaptogenic.
Immunostaining of RGCs for co-localization of presynaptic synaptogamin and
postsynaptic
PSD-95 shows few synaptic punta in the absence of astrocytes (B), but many in
the presence of
astorcyte feeding layers inserts (+ Astrocyte) or TSPs 3, 4, and 5 mimic the
synaptogenic effect
of astrocytes or TSPI and increase co-localized synaptic puncta.
[0027] Figures 1G: Pentameric Subgroup B Thrombospondins (TSPs) are
Synaptogenic.
Quantification of the effects of astrocytes, purified TSP1, and purified
recombinant TSPs 4 and 5
(8nM each) on synapse number. TSPs 4 and 5 increased synaptic puncta numbers
to the same
extent as either astrocytes or TSP1, indicating the TSPs 4 and 5 are also
synaptogenic.
[0028] Figures IH: Pentameric Subgroup B Thrombospondins (TSPs) are
Synaptogenic.
Quantification of the effects of astrocytes, and overexpressed TSP3 on number
of synapses made
by RGCs. TSP3 was overexpressed in Cos7 cells and RGCs were fed with this Cos7
cell culture
supematant. As a control condition, RGCs were fed with cell culture
supernatant from Cos7 cells
transfected with the empty vector. TSP3-containing culture supernatant
increased the number of
synaptic puncta to the same extent as astrocytes, indicating TSP3 is also
synaptogenic
[0029] Figures 2A: The EGF-like domains mediate thrombospondin's synaptogenic
effect.
The domain structure of TSPI and 2. TSPs 1 and 2 contain a heparin binding N-
terminal domain
(N), followed by an oligomerization domain and a procollagen repeat (PC),
three properdin-like
(TSP type 1), three EGF-like (TSP type 2), and thirteen calcium binding (TSP
type 3) repeats
and a C-terminal L-type lectin like globular domain (C).
[0030] Figures 2B: The EGF-like domains mediate thrombospondin's synaptogenic
effect.
Quantification of the effect of purified TSP 1 truncation constructs on
synapse number. RGCs
were treated with astrocytes, full-length TSP1 or a panel of TSPI truncation
constructs (8nM
each). The constructs that contained the EGF-like repeats of TSP1 were
synaptogenic.
[0031] Figures 2C: The EGF-like domains mediate thrombospondin's synaptogenic
effect.
Quantification of the effect of TSP2 truncation constructs on synapse number.
Similar to TSPI
constructs the TSP2 fragments that contained the EGF-like repeats were also
synaptogenic. A
construct that contained the third EGF-like domain attached to the C-terminal
region of the

7


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
protein still retained most of its synaptogenic activity, however the third
EGF-like domain alone
did not increase the number of synapses significantly.
[0032] Figures 2D-E: The EGF-like domains mediate thrombospondin's
synaptogenic
effect. Antibodies against EGF like repeats of TSPs can block their
synaptogenic effect. RGCs
cultured with astorcytes or with the recombinant TSP1 (Figure 2D) or TSP2
(Figure 2E)
truncation constructs that contained the third properdin repeats with the
three EGF-like domains
formed many more synapses when compared to RGCs cultured alone.
[0033] Figures 3A-E: Gabapentin blocks TSP induced synapse formation in vitro.
[0034] Figure 4A: Calcium channel subunit a261 is the TSP receptor involved in
synaptogenesis. RGCs were transfected with empty vector (pcDNA3, Invitrogen)
or a vector that
expresses a281 (pcDNA3-(x26 1). The synapses received by transfected cells
(marked by GFP
co-expression) were then quantified. In RGCs transfected with an empty vector
the cells
responded to the synaptogenic domain of TSP (SD2) by increasing the number of
synapses
formed over 5 fold. Overexpression of a281 enhanced the ability of TSP to
induce synapse
number 2 fold. n=20 cells, Error bars mean SEM, * p<0.05.

[0035] Figure 4B: Calcium channel subunit a281 is the TSP receptor involved in
synaptogenesis. a28l expression can be knocked down by a specific siRNA pool
(Dharmacon)
directed against a281 mRNA. Western blot analysis of cell lysates from HEK293
cells, which
were co-transfected with an expression vector for rat a281 and siControl or
sia28l pools, with a
monoclonal antibody against a281 showed that a281 expression was specifically
knocked down
by the sia281 pool. Same samples were blotted with an antibody against 0-actin
that served as
the loading control.
[0036] Figure 4C: Calcium channel subunit a281 is the TSP receptor involved in
synaptogenesis. Immunostaining of siRNA transfected RGCs (marked blue by GFP
co-
expression) for co-localization of presynaptic synaptotagmin and postsynaptic
PSD-95. RGCs
form few synaptic puncta in the absence of astrocytes (alone), but many in the
presence of
astrocytes when they are transfected with the non-targeting siControl pool. On
the other hand
RGCs that were transfected with sia281 did not form many synapses even in the
presence of
astrocytes. Scale bars=30 m.

8


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
[0037] Figure 4D: Calcium channel subunit a281 is the TSP receptor involved in
synaptogenesis. Quantification of the effects of siRNA pools on astrocyte-
induced synapse
formation in RGCs. RGCs transfected with the non-targeting siControl pool or
with a targeting
siRNA pool against another TSP receptor integrin (31 (silnt(31) still formed
many synapses in
response to astrocytes, however in RGCs transfected with sia281 pool,
astrocyte-induced
synapse formation was inhibited. n=20 cells, Error bars mean SEM, * p<0.05.

[0038] Figure 4E: Calcium channel subunit a261 is the TSP receptor involved in
synaptogenesis. Quantification of the effects of Sl overexpression on TSP-
induced synapse
formation in RGCs. RGCs that were transfected with a vector expressing only
the 81 portion of
the a281 were assayed for TSP-induced synapse formation. The synapses received
by
transfected cells were quantified. RGCs transfected with a vector expressing
GFP alone
responded to the synaptogenic domain of TSP (SD2) by increasing the number of
synapses
formed significantly, however the overexpression of 81 blocked the ability of
SD2 to induce
synapse formation. n=20 cells, Error bars mean SEM, * p<0.05.
[0039] Figure 5A. Calcium channel function or expression levels do not effect
TSP/astrocyte induced synapse formation in vitro. Quantification of the effect
of L-type calcium
channel blockers nimodipine and nifedipine (Sigma) on the synaptogenic
activity of SD2. SD2
was able to induce a significant increase in synapse number even in the
presence of nifedipine or
nimodipine (4 and 0.5 M, respectively) indicating that L-type calcium channel
function is not
required for the synaptogenic function of TSP. n=20 cells, Error bars mean
SEM, * p<0.05.
[0040] Figure 5B. Calcium channel function or expression levels do not effect
TSP/astrocyte induced synapse formation in vitro. Quantification of the effect
of overexpression
of L-type calcium channel subunits a 1 C and 0 in astrocyte induced synapse
formation.
Overexpression of a1C and (3 subunits did not have any positive or negative
effect on the
astrocytes ability to induce synapse formation. n=20 cells, Error bars mean
SEM, * p<0.05.
[0041] Figure 6A: a251 overexpression in vivo increases excitatory synapse
number.
Cortices from littermate wildtype (WT) and a281 overexpressing transgenic (TG)
P21 mice
were immunolabeled for pre-synaptic VGlut2 and post-synaptic PSD95. Number of
co-localized
VGlut2/PSD95 puncta (white arrows in inlays i and ii) was noticeably higher in
the TGs then the
WTs. Scale bars=20 m.

9


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
[0042] Figure 6B: a281 overexpression in vivo increases excitatory synapse
number.
Quantification of co-localization of pre- and post-synaptic markers VGlut2 and
PSD95 in brain
sections from WT and TG mice. TG brains showed around a 1.6 fold increase in
the number of
VGlut2-PSD95 synapses when compared to WT brains (*p<0.05).
[0043] Figure 6C: a281 overexpression in vivo increases excitatory synapse
number.
Cortices from WT and TG mice were also immunolabeled for VGlutl and PSD95.
Numbers of
co-localized VGlutl/PSD95 puncta (white arrows in inlays i and ii) were
similar in the TGs and
the WTs. Scale bars = 20 m.
[0044] Figure 6D: a261 overexpression in vivo increases excitatory synapse
number.
Quantification of co-localization of pre- and post-synaptic markers VGlutl and
PSD95 in brain
sections from WT and TG mice. Similar numbers of co-localized VGlutl/PSD95
synapses were
observed in TG brains compared to WT brains.
[0045] Figure 7A: Gabapentin inhibits TSP/astrocyte induced synapse formation.
Immunostaining of RGCs for co-localization of presynaptic synaptotagmin (red)
and
postsynaptic PSD-95 shows few synaptic puncta when RGCs are cultured alone,
but many in the
presence of SD2. Addition of GBP at 32 M concentration from the beginning of
the SD2
treatment inhibited TSP induced synapse formation visualized by the lack of co-
localized pre and
post-synaptic puncta in SD2 plus GBP condition (inlays i versus ii). Scale
bars = 30 m.
[0046] Figure 7B: Gabapentin inhibits TSP/astrocyte induced synapse formation.
Quantification of the effect of GBP on SD2-induced synapse formation. GBP
blocks TSPs
synaptogenic effect only when added to the RGCs at the same time as the
synaptogenic domain
SD2. It does not decrease synapse numbers when added to the cells for the last
24 hours. n=20
cells, Error bars mean SEM, * p<0.05.
[0047] Figure 7C: Gabapentin inhibits TSP/astrocyte induced synapse formation.
Quantification of the effect of GBP on astrocyte-induced synapse formation.
RGCs were treated
either with rat or mouse ACM in the presence or absence of 32 M GBP. GBP is
able to block
all of ACMs synaptogenic activity. n=20 cells, Error bars mean SEM, *
p<0.05.
[0048] Figure 7D: Gabapentin inhibits TSP/astrocyte induced synapse formation.
Quantification of co-localization of pre- and postsynaptic markers VGlut2 and
PSD95 in brain
sections from saline and GBP injected P7 mice. GBP injected brains showed a
significant



CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
decrease in the number of VGlut2-PSD95 synapses when compared to saline
injected brains
(*p<0.05).
[0049] Figure 7E: Gabapentin inhibits TSP/astrocyte induced synapse formation.
Saline and
GBP injected P7 cortices were immunolabeled for presynaptic VGlut2 and
postsynaptic PSD95.
In 50% of the GBP injected mice there was a very severe reduction in the
number, size and co-
localization of synaptic puncta containing both pre- and postsynaptic markers
VG1ut2 and
PSD95 (white arrows, inlays i versus ii). Scale bars = 20 m.
[0050] Figure 8: GABA mimics the effect of GBP in inhibiting TSP induced
synaptogenesis in culture. Quantification of the effect of GABA on SD2 induced
synapse
formation. GBP (32 M) blocks the synaptogenic activity of SD2 as shown
before. GABA was
also inhibitory when it was added to RGCs at 1mM concentration. GABA did not
decrease the
synapse numbers induced by SD2 when it was used at lower (10 and 100 M)
concentrations.
n=20 cells, Error bars mean SEM, * p<0.05.
[0051] Figure 9A: TSP induced synapse formation is involved in barrel cortex
plasticity.
The organization of the whisker pad is recapitulated point to point in the
barrel cortex of the
mouse. Schematic of the experimental paradigm: ablation of the C-row of
whiskers at P1 causes
corresponding contralateral barrel representations at P7 to shrink and fuse,
while neighboring
barrels invade the territory of deprived barrels.
[0052] Figure 9B: TSP induced synapse formation is involved in barrel cortex
plasticity.
Immunostaining in tangential cortical sections with an antibody against the
serotonin transporter
(5-HTT) labels thalamocortical afferents to the barrel cortex. Left images
show barrel cortex
contralateral to intact whisker pad (unlesioned "control" side). Right images
are the
representative examples of lesion-induced plasticity following whisker
follicle ablation in mice
that were injected with saline (top), with GBP (middle). Bottom row are the
control (left) and
lesioned (right) barrel cortices from a TSP1/2K0 mouse. Arrows flank the C-row
of barrels
corresponding to lesioned whiskers. Brackets and dashed lines show the
expansion of D-row
barrels. Asterisks denote regions of abnormal lesion-induced plasticity.
[0053] Figure 9C: TSP induced synapse formation is involved in barrel cortex
plasticity.
Hematoxylin staining of the whisker pads from the same mice whose barrels are
shown in (B)
showing the lack of C row while he neighboring rows of follicles are still
present.

11


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
[0054] Figure 10: A collection of the barrel cortex plasticity phenotypes in
saline and GBP
injected mice. Immunostaining in tangential cortical sections with an antibody
against the
serotonin transporter (5-HTT) labels thalamocortical afferents to the barrel
cortex. For each
mouse labeled 1-8, left images show barrel cortex contralateral to intact
whisker pad
(unlesioned-control side). Right images are the representative examples of
lesion-induced
plasticity following whisker follicle ablation in mice that were injected with
saline (mice 1-4),
with GBP (mice 5-8). GBP injection did not affect the barrel cortex formation
as the barrels
formed in a similar fashion to saline injected ones (mice 1-8 left panels),
however, in 50% of the
mice GBP injection induced aberrant barrel cortex plasticity phenotypes. In
these mice typically
the A, B, and C rows of the lesioned cortex lost form diffused and merged
together (mouse 5). In
one case only the neighboring D row merged and diffused with the C row and
lost form. In one
extreme case the majority of the barrel cortex had lost its shape and was
diffused (mouse 7).
None of the mice injected with saline ever showed these kinds of phenotypes
and they all
displayed normal barrel cortex plasticity (mice 1- 4, right panels). Half of
the GBP injected
animals also displayed a normal barrel cortex plasticity phenotype (mice 8).
[0055] Figure 11A: Thrombospondins interact with calcium channel subunit a281.
RGCs
express calcium channel subunit a281 in culture. Nine DIV RGC lysates were
analyzed by
Western blotting for the presence of a281 protein using a monoclonal antibody
against a28 I.
RGCs expressed a281 when they were cultured alone (lane 1) or in the presence
of astrocytes
(lane 2) or in the presence of TSP1 (lane 3). a281 levels did not change with
any of these
treatments. (3-actin level in each sample was tested to serve as a loading
control.

[0056] Figure 11B: Thrombospondins interact with calcium channel subunit a28
1. a261
immunoprecipitates with TSP 1, 2 and 4 from P5 rat brain cortical lysate. TSPs
1, 2 and 4 were
immunoprecipitated from P5 cortical lysates using specific rabbit polyclonal
antibodies. a281
was detected by Western-blot analysis on the immunoprecipitation (IP)
fractions. TSPs1, 2 or 4
co-immunoprecipitated a281, whereas no a251 was detected in the IP fraction of
a mock rabbit
polyclonal antibody.
[0057] Figure 11 C: Thrombospondins interact with calcium channel subunit a28
1. a251
interacts with the synaptogenic domain of TSP2 (SD2). FLAG tagged a251 was
expressed in
HEK293 cells alone (1) or in the presence of either SD2 (2) or another
unrelated secreted protein

12


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
Control-myc-his (3). a261 was then immunoprecipitated from HEK293 cell
membrane lysates
(lanes 1 through 3) using agarose beads conjugated to an antibody against the
FLAG-tag.
Presence of SD2 or Control-myc-his protein in the IP fractions was analyzed by
Western blotting
using anti-His-tag antibody (Lanes 4-6). SD2 co-immunoprecipitated with a281
(lane 5), while
the control-his-myc protein did not (lane 6) demonstrating a specific
interaction between SD2
and a2S 1.
[0058] Figure 11 D: Thrombospondins interact with calcium channel subunit
a261. SD2
interacts with a2S1, but not with the calcium channel subunit a1C. SD2, and
calcium channel
subunits al, a281 and 0 were co-expressed at different combinations in HEK 293
cells. Lane 1:
a281, a1C and 0 subunits of L-type calcium channel. Lane 2: a1C and (3
subunits and SD2.
Lane 3: a261 and SD2. Lane 4: a 1 C, a281 and (3 subunits and SD2. SD2 protein
was
immunoprecipitated from these solubilized HEK293 membranes by utilizing its C-
terminal myc-
tag. Presence of calcium channel subunits a281 or alC in the IP fractions were
analyzed. SD2
and a281 were co-immunoprecipitated regardless of the absence or presence of
a1 and (3
subunits (lanes 7 and 8). SD2 did not interact with a 1 C in the presence or
absence of the a251
subunit (lanes 6 and 8).
[0059] Figure 12A: Preparation and testing of the TSP2 synaptogenic domain
construct
SD2. Schematic representation of the synaptogenic TSP2 construct SD2. This
construct
contains a signal sequence for secretion from IgG K chain (SS) followed by the
third properdin
like repeat (P3) and the three synaptogenic EGF-like repeats (E1-3) of TSP2. C-
terminal myc-
and His- tags were introduced for immunodetection, immunoprecipitation and
purification
purposes.
[0060] Figure 12B: Preparation and testing of the TSP2 synaptogenic domain
construct
SD2. SD2 was produced in high amounts by overexpressing in HEK293 cells. The
protein was
then purified to homogeneity from culture media by using Ni-chelating
chromatography. Column
elution fractions that contain the pure protein were analyzed by SDS-PAGE and
the purity of the
protein was checked by commassie staining (lanes 1-3). M=molecular weight
marker, 206, 130,
87, 42, 31, 17, and 7 kDa (BioRad).
[0061] Figure 12C: Preparation and testing of the TSP2 synaptogenic domain
construct
SD2. Immunostaining of RGCs for colocalization of presynaptic synaptotagmin
and postsynaptic
13


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
PSD-95 shows few synaptic puncta when RGCs are cultured alone (Control), but
many in the
presence of SD2 (white arrows in inlay ii). Scale bars = 30 m.
[0062] Figure 12D: Preparation and testing of the TSP2 synaptogenic domain
construct
SD2. Quantification of the dose response for the effect of SD2 on synapse
number. SD2 was
most effective in increasing the synapse number at 10-20 nM range, but lost
its synaptogenic
activity above 50 nM concentration. N=20 cells, error bars mean + SEM, *
p<0.05.
[0063] Figure 13A: Intrathecal injection of TSP4 proteins into naive rats
induced behavioral
hypersensitivity in a dose-dependent and reversible manner. The onset time of
the TSP protein
effects was 2 days post injection, and the peak effects occurred 2-4 days post
injection.
Gabapentin or saline bolus treatment injection started three days after the
TSP4 bolus injection
(45 gg/rat), followed by behavioral test 1 hr after the treatment injection,
and further followed by
daily behavioral test. Intrathecal bolus gabapentin, but not saline, blocked
the pain-inducing
effects of TSP4. The gabapentin effects lasted for 1-2 days. PWT means paw
withdrawal
thresholds to von Frey filament stimulation.
[0064] Figure 13B-C: Intrathecal injection of active TSP4 antibody reversed
tactile
allodynia in spinal nerve ligated rats in a dose-dependent manner. Bolus
intrathecal TSP4
antibody at 80 gg/rat or at various doses were injected into two-week L5/6
spinal nerve ligated
rats, followed by von Frey test (Fig 13B-C). The bolus active TSP4 antibody
reversed
established allodynia at the injury site. In Figure 13B and C, "PWT" refers to
paw withdrawal
thresholds to von Frey filament stimulation; "Active" refers to active
antibody; "Inactive" refers
to antibody boind for 10 min.; "Contra" refers to non-injury side; "Ips"
refers to injury side; error
bars mean SEM; and "*"indicates p<0.05 and "**" indicates p<0.01 as compared
to pre-
treatment level.
[0065] Figure 13D: Pre-emptive intrathecal injection of TSP4 antisera
prevented the
development of tactile allodynia in spinal nerve ligated rats. Preemptive
intrathecal daily
treatment with TSP4 antibody (80 g/rat/day) started prior to the ligation of
the left L5/6 spinal
nerve. The preemptive treatment with TSP4 antibody delayed the onset of injury-
induced
allodynia shown as reduced paw withdrawal thresholds (PWT) to mechanical
stimulation. Data
shown are the Mean + SEM. "L" refers to left (ligation) side; and "R" refers
to right (non-injury)
side. "*" indicates p<0.05 as compared to saline (L); "**" indicates p<0.01 as
compared to
saline (L); and "***" indicates p < 0.001 as compared to saline (L).

14


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
[0066] Figure 13E: Intrathecal injection of TSP4 antisense
oligodeoxynucleotides reversed
tactile allodynia in the spinal nerve ligated rats in a sequence-specific and
reversible manner.
Two TSP4 antisense oligonucleotides (#1 and #2) were injected intrathecally
(50 g/rat/day) into
spinal nerve ligated rats for 4 days 5 weeks after ligation injury. Daily
intrathecal injection of
TSP4 antisense oligonucleotide #2 caused a complete reversal of established
allodynia in the
injury side. Daily behavioral test was performed before injection blindly.
Mean + SEM from n=
6 in each group is shown. "Contra" refers to non-injury side; and "Ips" refers
to L5/6 spinal
nerve ligated side. "*" indicates p<0.05 as compared to pre-treatment levels.

DETAILED DESCRIPTION OF THE INVENTION
[0067] Methods and compositions are provided for protecting or treating an
individual
suffering from or preventing an individual from adverse effects of deficits in
synaptogenesis, or
from undesirably active synaptogenesis to a individual in need thereof. These
findings have
broad implications for a variety of clinical conditions, including traumatic
brain injury, epilepsy,
and other conditions where synapses fail to form or form inappropriately.
Synaptogenesis is
enhanced by contacting neurons with agents that are specific agonists of
thrombospondins.
Conversely, synaptogenesis is inhibited by contacting neurons with inhibitors
or antagonists of
thrombospondins.
[0068] Delivery of an exogenous thrombospondin or an agonist thereof induces
new
synapses in normal CNS, after CNS injury to promote repair, at neuromuscular
junctions, e.g. at
the junctions of spinal motor neurons and muscles. The ability to restore
synaptogenesis in an
adult has important implications for enhancing memory in normal brain; for
treatment of
Alzheimer's disease (a disease where synapses are lost), as well as promoting
new
synaptogenesis in repair and regeneration of injured CNS after stroke or
spinal cord injury;
enhancement of neuromuscular junctions in muscular dystrophy; amyotrophic
lateral sclerosis
(ALS); and the like. Delivery of an exogenous thrombospondin or an agonist
thereof also find
use in combination with administration of neural progenitors, or increases in
neurogenesis, in
order to promote functional connections between the nascent neurons and other
neurons and
effector cells.
[0069] Thrombospondin antagonists are useful in treating diseases of excess,
unwanted
synapses. The adult brain may upregulate thrombospondin after injury in
"reactive astrocytes",
which form glial scars. Glial scars are associated with epileptic loci, and
may induce the



CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
unwanted excess synaptogenesis that underlies epilepsy. Similarly there are
unwanted extra
synapses that underlie the long-lived drug craving of addiction and/or pain.
[0070] Methods are provided for the modulation of synaptogenesis with soluble
factors. It
has been found that thrombospondin is sufficient to increase synapse formation
on neurons, in
particular synapse formation is increased by the action of the common EGF like
domain shared
by thrombospondins, including the third EGF-like domain. This domain interacts
with a widely
expressed transmembrane neuronal cell surface molecule, calcium channel
subunit a281. Agents
that block the interaction between thrombospondin and a281 are useful in
inhibiting synapse
formation, e.g. in the treatment of pain, epilepsy, anxiety, addiction, and to
aid in the axon
growth of regenerating neurons. It has been found that Gabapentin, which binds
to a281, and
antibodies specific to the thrombospondin EGF-like domain, specifically
inhibit synapse
formation induced by thrombospondin. Methods of interest include the
enhancement of
synaptogenesis by contacting neurons with a thrombospondin EGF-like domain, or
mimetic
thereof such as agonistic antibodies, and the inhibition of synaptogenesis by
blocking the
interaction between thrombospondin EGF-like domain and a a281, a252, a283, or
a264
polypeptide or protein.
[0071] In one embodiment of the invention, methods are provided for screening
candidate
agents for the ability to modulate synapse formation, including the inhibition
of synapse
formation. In one embodiment of the invention, the neurons are neurons in the
central nervous
system. In another embodiment, the neurons are peripheral nervous system
neurons. Screening
may include contacting a a281, a262, a283, or a264 polypeptide or protein, and
determining the
ability of an agent to bind to, or otherwise interact with a281, a262, a263,
or a284. Such
screening assays may further include determining the effect of a candidate
agent on cells
expressing a25 1, a262, a283, or a264. Such agents are candidate for
therapeutic treatment of
epilepsy and other conditions characterized by undesirable synaptogenesis.
[0072] Thrombospondin, agonists, and mimetics thereof, are administered to
enhance
synaptogenesis, particularly thrombospondin peptides comprising at least an
EGF-like domain or
agonistic antibodies that mimic the activity of the thrombospondin EGF-like
domain. All 5
known thrombospondin isoforms have strong synapse inducing activity as a
result of sharing the
EGF-like domain. Inhibitors, e.g. antibodies, gabapentin and analogs thereof,
etc. are
administered to inhibit synaptogenesis.

16


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
[0073] The synaptogenic domain of TSP is useful in stimulating synapse
formation in areas
where synapses are lost due to neurodegenerative diseases, e.g. Alzheimer's
disease, Parkinson's
disease, ALS, multiple sclerosis, retinal degeneration, glaucoma, stroke,
neuropathy aging, etc.
In other embodiments, agent that block TSP mediated synapse formation, e.g.
a281 ligands such
as gabapentin, pregabalin, etc.; antibodies specific for the EGF-like domain
of TSP, etc. are
useful in inhibiting synapse formation. Such methods find use in preventing
aberrant synapse
formation after injury to a regenerating axon, e.g. post injury to nerves
including optic nerve, in
glaucoma or spinal cord neurons following spinal cord injury, etc. The
synaptogenesis
modulators may be administered topically, e.g. to optic nerves or spinal cord.
Definitions
[0074] Synaptogenesis, as used herein, refers to the process by which pre-
and/or post-
synapses form on a neuron. Enhancing synaptogenesis results in an increased
number of
synapses, while inhibiting synaptogenesis results in a decrease in the number
of synapses, or a
lack of increase where an increase would otherwise occur. By "augmentation" or
"modulation"
of synaptogenesis as used herein, it is meant that the number of synapses
formed is either
enhanced or suppressed as required in the specific situation.
[0075] As used herein, the term "thrombospondin" may refer to any one of the
family of
proteins which includes thrombospondins I, II, III, IV, and cartilage
oligomeric matrix protein.
Reference may also be made to one or more of the specific thrombospondins.
Thrombospondin
is a homotrimeric protein composed of three identical subunits (TSP 1 and
TSP2) or
homopentameric protein composed of five identical subunits (TSPs 3-5)
glycoprotein with
disulfide-linked subunits of MW 180,000. It contains binding sites for
thrombin, fibrinogen,
heparin, fibronectin, plasminogen, plasminogen activator, collagen, laminin,
calcium etc. and
also contains domain homologues to procollagen, properdin, and epidermal
growth factor (EGF).
It functions in many cell adhesion and migration events, including platelet
aggregation.
[0076] As used herein, the term "modulator of synaptogenesis" refers to an
agent that is
able to alter synapse formation. Modulators include, but are not limited to,
both "activators" and
"inhibitors". An "activator" or "agonist" is a substance that enhances
synaptogenesis.
Conversely, an "inhibitor" or "antagonist" decreases the number of synapses.
The reduction may
be complete or partial. As used herein, modulators encompass thrombospondin
antagonists and
agonists.

17


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
[0077] As used herein, the term "modulator of axonal and/or dendritic growth"
refers to an
agent that is able to alter axonal and/or dendritic growth. Modulators
include, but are not limited
to, both "activators" and "inhibitors". An "activator" or "agonist" is a
substance that enhances
axonal and/or dendritic growth. Conversely, an "inhibitor" or "antagonist"
decreases axonal
and/or dendritic growth. The reduction may be complete or partial. As used
herein, modulators
encompass thrombospondin antagonists and agonists.
[0078] As used herein, "agent" refers to a biological, pharmaceutical, or
chemical
compound. Non-limiting examples include simple or complex organic or inorganic
molecule, a
peptide, a protein, an oligonucleotide, an antibody, an antibody derivative,
antibody fragment, a
vitamin derivative, a carbohydrate, a toxin, or a chemotherapeutic compound.
Various
compounds can be synthesized, for example, small molecules and oligomers
(e.g., oligopeptides
and oligonucleotides), and synthetic organic compounds based on various core
structures. In
addition, various natural sources can provide compounds for screening, such as
plant or animal
extracts, and the like. A skilled artisan can readily recognize that there is
no limit as to the
structural nature of the agents of the present invention.
[0079] Agents that are employed in the methods of this invention can be
randomly selected
or rationally selected or designed. As used herein, in some embodiments, an
agent is said to be
randomly selected when the agent is chosen randomly without considering the
specific sequences
involved in the association of thrombospondin with a calcium channel (e.g.,
a1S2 calcium
channel). An example of randomly selected agents is the use of a chemical
library or a peptide
comb inatori al library.
[0080] Agonists and antagonists may include proteins (i.e., polypeptides),
nucleic acids,
carbohydrates, antibodies, or any other molecules that affects a protein
and/or molecule of
interest. In some embodiments, an antagonist may inhibit (e.g., decrease) one
or more activities
or functions of a protein and/or molecule of interest. In some embodiments, an
agonist may
stimulate (e.g., increase) one ore more activities or functions of a protein
and/or molecule of
interest. The term "analog" is used herein to refer to a molecule that
structurally or functionally
resembles a molecule of interest but which has been modified in a targeted and
controlled
manner, by replacing a specific substituent of the reference molecule with an
alternate
substituent. Compared to the starting molecule, an analog may exhibit the
same, similar, or
improved utility. Synthesis and screening of analogs, to identify variants of
known compounds

18


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
having improved traits (such as higher potency at a specific receptor type, or
higher selectivity at
a targeted receptor type and lower activity levels at other receptor types) is
an approach that is
well known in pharmaceutical chemistry.
[0081] An "antibody" is an immunoglobulin molecule capable of specific binding
to a
target, such as a carbohydrate, polynucleotide, lipid, polypeptide, etc.,
through at least one
antigen recognition site, located in the variable region of the immunoglobulin
molecule. As used
herein, the term encompasses not only intact polyclonal or monoclonal
antibodies, but also
fragments thereof (such as Fab, Fab', F(ab')2, Fv), single chain (ScFv),
mutants thereof, naturally
occurring variants, fusion proteins comprising an antibody portion with an
antigen recognition
site of the required specificity, humanized antibodies, chimeric antibodies,
and any other
modified configuration of the immunoglobulin molecule that comprises an
antigen recognition
site of the required specificity.
[0082] The term "monoclonal antibody" encompasses not only intact monoclonal
antibodies and full-length monoclonal antibodies, but also fragments thereof
(such as Fab, Fab',
F(ab')2, Fv), single chain (ScFv), mutants thereof, fusion proteins comprising
an antibody
portion, humanized monoclonal antibodies, chimeric monoclonal antibodies, and
any other
modified configuration of the immunoglobulin molecule that comprises an
antigen recognition
site of the required specificity and the ability to bind to an antigen. It is
not intended to be limited
as regards to the source of the antibody or the manner in which it is made
(e.g., by hybridoma,
phage selection, recombinant expression, transgenic animals, etc.).
[0083] "Humanized" antibodies refer to a molecule having an antigen- binding
site that is
substantially derived from an immunoglobulin from a non-human species and the
remaining
immunoglobulin structure of the molecule based upon the structure. and /or
sequence of a human
immunoglobulin. The antigen-binding site may comprise either complete variable
domains fused
onto constant domains or only the complementarity determining regions (CDRs)
grafted onto
appropriate framework regions in the variable domains. Antigen binding sites
may be wild type
or modified by one or more amino acid substitutions, e.g., modified to
resemble human
immunoglobulin more closely. Some forms of humanized antibodies preserve all
CDR sequences
(for example, a humanized mouse antibody which contains all six CDRs from the
mouse
antibodies). Other forms of humanized antibodies have one or more CDRs (one,
two, three, four,

19


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
five, six) which are altered with respect to the original antibody, which are
also termed one or
more CDRs "derived from" one or more CDRs.
[0084] "Chimeric antibodies" refers to those antibodies wherein one portion of
each of the
amino acid sequences of heavy and light chains is homologous to corresponding
sequences in
antibodies derived from a particular species or belonging to a particular
class, while the
remaining segment of the chains is homologous to corresponding sequences in
another.
Typically, in these chimeric antibodies, the variable region of both light and
heavy chains
mimics the variable regions of antibodies derived from one species of mammals,
while the
constant portions are homologous to the sequences in antibodies derived from
another. One clear
advantage to such chimeric forms is that, for example, the variable regions
can conveniently be
derived from presently known sources using readily available hybridomas or B
cells from non
human host organisms in combination with constant regions derived from, for
example, human
cell preparations. While the variable region has the advantage of ease of
preparation, and the
specificity is not affected by its source, the constant region being human, is
less likely to elicit an
immune response from a human subject when the antibodies are injected than
would the constant
region from a non-human source. However, the definition is not limited to this
particular
example.
[0085] As used herein, the term "immunoadhesin" designates antibody-like
molecules
which combine the binding specificity of a heterologous protein (an
"adhesion") with the
effector functions of immunoglobulin constant domains. Structurally, the
immunoadhesins
comprise a fusion of an amino acid sequence with the desired binding
specificity which is other
than the antigen recognition and binding site of an antibody (i.e., is
"heterologous"), and an
immunoglobulin constant domain sequence. The adhesin part of an immunoadhesin
molecule
typically is a contiguous amino acid sequence comprising at least the binding
site of a receptor or
a ligand. The immunoglobulin constant domain sequence in the immunoadhesin may
be obtained
from any immunoglobulin, such as IgG-1, IgG-2, IgG-3, or IgG-4 subtypes, IgA
(including IgA-
1 and IgA-2), IgE, IgD or IgM. The Ig fusions preferably include the
substitution of a domain of
a polypeptide or antibody described herein in the place of at least one
variable region within an
Ig molecule. In a particularly preferred embodiment, the immunoglobulin fusion
includes the
hinge, CH2 and CH3, or the hinge, CH1, CH2 and CH3 regions of an IgGI
molecule. For the
production of immunoglobulin fusions see also US Patent No. 5,428,130 issued
June 27, 1995.



CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
In an embodiment, the invention provides an immunoadhesion comprising all or a
part of the
extracellular domain of a calcium channel (e.g., a28 subunit or a2 subunit),
or fragments thereof
and a fusion partner, such as an antibody Fc domain.
[0086] A "variable region" of an antibody refers to the variable region of the
antibody light
chain or the variable region of the antibody heavy chain, either alone or in
combination. The
variable regions of the heavy and light chain each consist of four framework
regions (FR)
connected by three complementarity determining regions (CDRs) also known as
hypervariable
regions. The CDRs in each chain are held together in close proximity by the
FRs and, with the
CDRs from the other chain, contribute to the formation of the antigen-binding
site of antibodies.
There are at least two techniques for determining CDRs: (1) an approach based
on cross-species
sequence variability (i.e., Kabat et al. Sequences of Proteins of
Immunological Interest, (5th ed.,
1991, National Institutes of Health, Bethesda MD)); and (2) an approach based
on
crystallographic studies of antigen-antibody complexes (Al-lazikani et al
(1997) J. Molec. Biol.
273:927-948)). As used herein, a CDR may refer to CDRs defined by either
approach or by a
combination of both approaches.
[0087] A "constant region" of an antibody refers to the constant region of the
antibody light
chain or the constant region of the antibody heavy chain, either alone or in
combination.
[0088] An antibody "specifically binds" or "preferentially binds" to a target
if it binds with
greater affinity, avidity, more readily, and/or with greater duration than it
binds to other
substances. For example, an antibody that specifically or preferentially binds
to a
thrombospondin and/or calcium channel epitope is an antibody that binds this
thrombospondin
and/or calcium channel epitope with greater affinity, avidity, more readily,
and/or with greater
duration than it binds to other thrombospondin and/or calcium channel epitopes
or non-
thrombospondin and/or non-calcium channel epitopes. It is also understood by
reading this
definition that, for example, an antibody (or moiety or epitope) that
specifically or preferentially
binds to a first target may or may not specifically or preferentially bind to
a second target. As
such, "specific binding" or "preferential binding" does not necessarily
require (although it can
include) exclusive binding. Generally, but not necessarily, reference to
binding means
preferential binding.
[0089] As used herein, the term "labeled", with regard to the antibody, is
intended to
encompass direct labeling of the antibody by coupling (i.e., physically
linking) a detectable
21


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
substance, such as a radioactive agent or a fluorophore (e.g. fluorescein
isothiocyanate (FITC) or
phycoerythrin (PE)) to the antibody, as well as indirect labeling of the probe
or antibody by
reactivity with a detectable substance.
[0090] As used herein, "substantially pure" refers to material that is at
least 50% pure (i.e.,
free from contaminants), more preferably at least 90 % pure, more preferably
at least 95% pure,
more preferably at least 98% pure, more preferably at least 99% pure.
[0091] The terms "polypeptide", "oligopeptide", "peptide" and "protein" are
used
interchangeably herein to refer to polymers of amino acids of any length. The
polymer may be
linear or branched, it may comprise modified amino acids, and it may be
interrupted by non-
amino acids. The terms also encompass an amino acid polymer that has been
modified naturally
or by intervention; for example, disulfide bond formation, glycosylation,
lipidation, acetylation,
phosphorylation, or any other manipulation or modification, such as
conjugation with a labeling
component. Also included within the definition are, for example, polypeptides
containing one or
more analogs of an amino acid (including, for example, unnatural amino acids,
etc.), as well as
other modifications known in the art. It is understood that, because the
polypeptides of this
invention are based upon an antibody, the polypeptides can occur as single
chains or associated
chains.
[0092] An "isolated" nucleic acid molecule encoding the antibodies herein is a
nucleic acid
molecule that is identified and separated from at least one contaminant
nucleic acid molecule
with which it is ordinarily associated in the environment in which it was
produced. Preferably,
the isolated nucleic acid is free of association with all components
associated with the production
environment. The isolated nucleic acid molecules encoding the polypeptides and
antibodies
herein is in a form other than in the form or setting in which it is found in
nature. Isolated nucleic
acid molecules therefore are distinguished from nucleic acid encoding the
polypeptides and
antibodies herein existing naturally in cells.
[0093] The term "control sequences" refers to DNA sequences necessary for the
expression
of an operably linked coding sequence in a particular host organism. The
control sequences that
are suitable for prokaryotes, for example, include a promoter, optionally an
operator sequence,
and a ribosome binding site. Eukaryotic cells are known to utilize promoters,
polyadenylation
signals, and enhancers.

22


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
[0094] Nucleic acid is "operably linked" when it is placed into a functional
relationship
with another nucleic acid sequence. For example, DNA for a presequence or
secretory leader is
operably linked to DNA for a polypeptide if it is expressed as a preprotein
that participates in the
secretion of the polypeptide; a promoter or enhancer is operably linked to a
coding sequence if it
affects the transcription of the sequence; or a ribosome binding site is
operably linked to a coding
sequence if it is positioned so as to facilitate translation. Generally,
"operably linked" means that
the DNA sequences being linked are contiguous, and, in the case of a secretory
leader,
contiguous and in reading phase. However, enhancers do not have to be
contiguous. Linking is
accomplished by ligation at convenient restriction sites. If such sites do not
exist, the synthetic
oligonucleotide adaptors or linkers are used in accordance with conventional
practice.
[0095] The term "epitope tagged" when used herein refers to a chimeric
polypeptide
comprising a polypeptide or antibody described herein fused to a "tag
polypeptide". The tag
polypeptide has enough residues to provide an epitope against which an
antibody can be made,
yet is short enough such that it does not interfere with activity of the
polypeptide to which it is
fused. The tag polypeptide preferably also is fairly unique so that the
antibody does not
substantially cross-react with other epitopes. Suitable tag polypeptides
generally have at least six
amino acid residues and usually between about 8 and 50 amino acid residues
(preferably,
between about 10 and 20 amino acid residues).
[0096] A "host cell" includes an individual cell or cell culture that can be
or has been a
recipient for vector(s) for incorporation of polynucleotide inserts. Host
cells include progeny of a
single host cell, and the progeny may not necessarily be completely identical
(in morphology or
in genomic DNA complement) to the original parent cell due to natural,
accidental, or deliberate
mutation. A host cell includes cells transfected in vivo with a
polynucleotide(s) of this invention.
[0097] As used herein, "percent (%) amino acid sequence identity" and
"homology" with
respect to a peptide, polypeptide or antibody sequence refers to the
percentage of amino acid
residues in a candidate sequence that are identical with the amino acid
residues in the specific
peptide or polypeptide sequence, after aligning the sequences and introducing
gaps, if necessary,
to achieve the maximum percent sequence identity, and not considering any
conservative
substitutions as part of the sequence identity. Alignment for purposes of
determining percent
amino acid sequence identity can be achieved in various ways that are within
the skill in the art,
for instance, using publicly available computer software such as BLAST, BLAST-
2, ALIGN or

23


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
MEGALIGNTM (DNASTAR) software. Those skilled in the art can determine
appropriate
parameters for measuring alignment, including any algorithms needed to achieve
maximal
alignment over the full length of the sequences being compared.
[0098] As used herein, the term "RNA interference" or "RNAi" refers generally
to a
process in which a double-stranded RNA molecule or a short hairpin RNA
molecule reducing or
inhibiting the expression of a nucleic acid sequence with which the double-
stranded or short
hairpin RNA molecule shares substantial or total homology. The term "short
interfering RNA" or
"siRNA" or "RNAi agent" refers to an RNA sequence that elicits RNA
interference. See
Kreutzer et al., WO 00/44895; Zernicka-Goetz et al., WO 01/36646; Fire, WO
99/32619; Mello
and Fire, WO 01/29058. As used herein, siRNA molecules include RNA molecules
encompassing chemically modified nucleotides and non-nucleotides. The term
"ddRNAi agent"
refers to a DNA-directed RNAi agent that is transcribed from an exogenous
vector. The terms
"short hairpin RNA" or "shRNA" refer to an RNA structure having a duplex
region and a loop
region. In certain embodiments, ddRNAi agents are expressed initially as
shRNAs.
[0099] "Carriers" as used herein include pharmaceutically acceptable carriers,
excipients, or
stabilizers that are nontoxic to the cell or mammal being exposed thereto at
the dosages and
concentrations employed. Often the physiologically acceptable carrier is an
aqueous pH buffered
solution. Examples of physiologically acceptable carriers include buffers such
as phosphate,
citrate, and other organic acids; antioxidants including ascorbic acid; low
molecular weight (less
than about 10 residues) polypeptide; proteins, such as serum albumin, gelatin,
or
immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino
acids such as
glycine, glutamine, asparagine, arginine or lysine; monosaccharides,
disaccharides, and other
carbohydrates including glucose, mannose, or dextrins; chelating agents such
as EDTA; sugar
alcohols such as mannitol or sorbitol; salt-forming counterions such as
sodium; and/or nonionic
surfactants such as TWEENTM, polyethylene glycol (PEG), and PLURONICSTM.
[0100] "Pharmaceutically acceptable" buffers and salts include those derived
from both acid
and base addition salts of the above indicated acids and bases. Specific
buffers and/ or salts
include histidine, succinate and acetate.
[0101] The term "pharmaceutical formulation" refers to a preparation that is
in such form as
to permit the biological activity of the active ingredient to be effective,
and that contains no

24


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
additional components that are unacceptably toxic to a subject to which the
formulation would be
administered. Such formulations are sterile.
[0102] By "neurological" or "cognitive" function as used herein, it is meant
that the
increase of synapses in the brain enhances the patient's ability to think,
function, etc. In
conditions where there is axon loss and regrowth, there may be recovery of
motor and sensory
abilities.
[0103] As used herein, the term "treatment" refers to clinical intervention
designed to alter
the natural course of the individual or cell being treated during the course
of clinical pathology.
Desirable effects of treatment include decreasing the rate of disease
progression, ameliorating or
palliating the disease state, and remission or improved prognosis. An
individual is successfully
"treated", for example, if one or more symptoms associated with the disease or
condition are
mitigated or eliminated.
[0104] As used herein, the term "prevention" includes providing prophylaxis
with respect to
occurrence or recurrence of a disease in an individual. An individual may be
predisposed to the
disease but has not yet been diagnosed with the disease.
[0105] An "effective amount" refers to at least an amount effective, at
dosages and for
periods of time necessary, to achieve the desired therapeutic or prophylactic
result. An effective
amount can be provided in one or more administrations.
[0106] A "therapeutically effective amount" is at least the minimum
concentration required
to effect a measurable improvement of a particular disorder. A therapeutically
effective amount
herein may vary according to factors such as the disease state, age, sex, and
weight of the patient,
and the ability of the antibody to elicit a desired response in the
individual. A therapeutically
effective amount is also one in which any toxic or detrimental effects are
outweighed by the
therapeutically beneficial effects.
[0107] A "prophylactically effective amount" refers to an amount effective, at
the dosages
and for periods of time necessary, to achieve the desired prophylactic result.
Typically but not
necessarily, since a prophylactic dose is used in subjects prior to or at an
earlier stage of disease,
a prophylactically effective amount may be less than a therapeutically
effective amount.
[0108] "Chronic" administration refers to administration of the medicament(s)
in a
continuous as opposed to acute mode, so as to maintain the initial therapeutic
effect (activity) for
an extended period of time.



CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
[0109] "Intermittent" administration refers to treatment that is not
consecutively done
without interruption, but rather is cyclic in nature.
[0110] As used herein, administration "in conjunction" includes simultaneous
administration and/or administration at different times. Administration in
conjunction also
encompasses administration as a co-formulation or administration as separate
compositions,
including at different dosing frequencies or intervals, and using the same
route of administration
or different routes of administration.
[0111] An "individual" or "subject" refers a mammal, including humans,
domestic and farm
animals, and zoo, sport, or pet animals, such as chimpanzees and other apes
and monkey species,
dogs, horses, rabbits, cattle, pigs, goats, sheep, hamsters, guinea pigs,
gerbils, mice, ferrets, rats,
cats, and the like. Preferably, the individual is human. The term does not
denote a particular age
or gender.
[0112] The term "about" as used herein refers to the usual error range for the
respective
value readily known to the skilled person in this technical field. Reference
to "about" a value or
parameter herein includes (and describes) embodiments that are directed to
that value or
parameter per se.
[0113] As used herein the singular forms "a", "and", and "the" include plural
referents
unless the context clearly dictates otherwise.
101141 It is understood that aspects and embodiments of the invention
described herein
include "consisting" and/or "consisting essentially of' aspects and
embodiments.

Methods of Modulating Axon Growth and/or Synaptogenesis
[0115] The present invention provides methods for modulating axonal growth
and/or
synaptogenesis in an individual. The methods comprise administering an
effective dose of an
agent which modulates to a thrombospondin and/or an a28 subunit of a calcium
channel (e.g., an
a26 1, a282, a263, and a284). In some embodiments, the agent binds to
thrombospondin and/or
an a28 subunit of a calcium channel. In some embodiments, the agent is an
agonist. In some
embodiments, the agent is an antagonist.
Calcium Channel, alpha-2/delta subunit
[0116] In some embodiments of any of the methods described herein, the calcium
channel is
a voltage-gated Caz+ (Cav) channels are composed of a pore-forming al subunit,
associated, at

26


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
least in the case of the Cavl and 2 subfamilies, with an intracellular (3
subunit responsible for
trafficking and a transmembrane a28 (such as a28 1) subunit. The al subunit
determines the main
biophysical properties of the channel and is modulated by the other subunits.
[0117] The CACNA2D 1 gene encodes the alpha-2/delta subunit of skeletal muscle
and
brain voltage-dependent calcium channels, which are heteromultimer complexes
comprising 4
subunits: alpha-1, alpha-2/delta, beta-1, and gamma. Alternative names for
this molecule include
alpha2delta Subunit 1; Cacna2dl; Calcium channel alpha2delta-1; Calcium
channel, voltage-
dependent, alpha2/delta 1 subunit; Calcium channel, voltage-dependent,
alpha2/delta subunit 1;
Cchl2a; and Voltage-dependent calcium channel alpha2delta-1. CACNA2D1 alters
the properties
of pore-forming alpha-1 subunits of voltage-gated calcium channels, and it is
posttranslationally
processed into 2 peptides, an alpha-2 subunit and a delta subunit, that are
held together by a
disulfide bond. The alpha-2/delta protein is encoded by at least 4 different
genes: CACNA2D 1
(a26 1), CACNA2D2 (a282), CACNA2D3 (a283), and CACNA2D4 (a284) (see, for
example
Schleithoff et al., 1999 Genomics 61: 201-209; and Field et al. (2006) Proc.
Nat. Acad. Sci. 103:
17537-17542, herein specifically incorporated by reference). The genetic
sequences and protein
sequences are publicly available. For example, genetic sequence for a261 is at
Genbank,
accession number BC117470; and protein sequence for a261 is at Genbank
accession number
AAI17471; protein sequence for a282 is at Genbank accession number AAI52439;
protein
sequence for a263 is at Genbank accession numbers AA137506 and AA137502; and
protein
sequence for a264 is at Genbank accession number AAI50187.
[0118] Iles et al. (1994) Hum. Molec. Genet. 3: 969-975 cloned and partially
sequenced the
CACNL2A gene. The CACNL2A is expressed in many tissues, including skeletal
muscle, brain,
heart, and lung. A comparison of sequences of cDNAs representing the skeletal
muscle and brain
isoforms showed that they are encoded by a single gene. The "delta" portion,
encoded by exons
37 to 40, is posttranscriptionally cleaved from the C-terminal "alpha" portion
of the protein. The
membrane-spanning region of the delta portion is encoded by exon 40. The
CACNA2D 1 gene
undergoes alternative splicing at exons 19 and 24, corresponding to muscle and
brain isoforms,
respectively.

[0119] The topology of the a28 protein appears to generalize for all four aZS
subunits. They
are all predicted to be type 1 transmembrane proteins, because all have a
hydrophobic region in
the C-terminus (CT) that is likely to be a transmembrane domain. a25 is
translated from a single
27


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
gene product, which gets post-translationally cleaved into a2 and S parts that
remain associated
via disulfide bridges. The a2 portion of the protein is entirely extracellular
while the S portion
has a small extracellular part that is attached to a2, and a transmembrane
domain with a very
short cytoplasmic tail that tethers the whole molecule to the membrane (Davies
et al., Trends in
Pharmacol. Sci. 28:220-228, 2007). All have predicted N-terminal signal
sequences, indicating
that the N terminus is extracellular. One domain identified by sequence
homology in the
extracellular sequence of all a26 subunits is the von Willebrand factor type A
(VWFA) domain
within the a2 moiety. Canti et al., Curr. Neuropharmacology 1:209-217, 2003;
Canti et al., Proc.
Natl. Acad. Sci. USA 102:11230-11235, 2005; Arikkath et al., Curr. Opin.
Neurobiol. 13,:298-
307, 2003. In human aZS subunits, VWFA domain comprises amino acids from about
253 to
about 430 of a281 (see worldwide web at expasy.org/uniprot/P54289), amino
acids from about
291 to about 469 of a282 (see worldwide web at expasy.org/uniprot/Q9NY47),
amino acids from
about 256 to about 438 of a283 (see worldwide web at
expasy.org/uniprot/Q8IZS8), and amino
acids from about 291 to about 473 of a264 (see worldwide web at
expasy.org/uniprot/Q7Z3S7).
The amino acid positions are based on the unprocessed precursor protein with
signal sequence.
[0120] All a28 subunits enhance calcium currents through the high-voltage-
activated (HVA)
Cavl and Cav2 channels. Voltage-activated Ca2+ channels are important
signaling proteins in
many cellular processes including muscle contraction, secretion, synaptic
function, and
transcriptional regulation. Expression of the a26-1 protein increases the
targeting of the al
subunit to the membrane and enhances gating of the pore-forming subunit (as
measured by
gating charge). In general, the co-expression of the a28-1 protein with the al
and 0 subunits of
the high-voltage-gated calcium channels shifts the voltage dependence of
activation and
inactivation to more negative potentials and accelerates the rates of channel
activation and
inactivation.
[0121] In addition to these direct actions on calcium channel function, the
a26-1 protein
mediates the actions of gabapentin and pregabalin, agents used in the
treatment of neuropathic
pain. In vitro studies have shown that this subunit is the binding site for
gabapentin, an
anticonvulsant that exerts antihyperalgesic effects. In vivo studies further
demonstrated that point
mutations in a25-1 protein eliminated the therapeutic effect of gabapentin in
a rodent model of
neuropathic pain. Increased expression of this subunit in the spinal cord and
dorsal root ganglia
(DRG) has been suggested to play a role in enhanced nociceptive responses of
spinal nerve-

28


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
injured rats to innocuous mechanical stimulation (allodynia). Induction of the
alpha2delta-1
subunit in the DRG and spinal cord is likely regulated by factors that are
specific for individual
neuropathies and may contribute to gabapentin-sensitive allodynia.
[0122] Gabapentin (1-(aminomethyl)cyclohexaneacetic acid) is considered by
physicians to
be the "gold standard" treatment for a variety of neuropathic pain. It is
prescribed to over 50% of
patients suffering from diabetic neuropathy or postherpetic neuralgia. The
drug is well tolerated
except for sedation seen at higher doses. Alternatively, the 3-substituted
analogue of y-
aminobutyric acid (GABA), pregabalin, provides for similar activity with an
improved
pharmacokinetic profile. Compounds represented by gabapentin and pregabalin
exert their effect
of blocking neuropathic pain by binding to the a281 subunit of voltage-gated
Ca2+ channel (see
Marais et al. (2001) Mol. Pharmacol. 59: 1243-1248 and Wang et al. (1999)
Biochem. J. 342:
313-320). This interaction was though to result in inhibition of calcium
influx into neuronal cells,
thereby inhibiting neurotransmitter release and suppressing the development of
central
sensitization.
[0123] The medicinal chemistry of a number of a261 ligands has been explored,
including
structural variants in the (3-amino acid and a-amino acid classes, non-amino
acid leads and
prodrugs. Binding of these ligands to the alpha2delta subunit is considered to
explain their
usefulness in treating several clinical disorders, including epilepsy, pain
from diabetic
neuropathy, postherpetic neuralgia and fibromyalgia, and generalized anxiety
disorder.
Thrombospondin
[0124] In some embodiments of any of the methods described herein,
"thrombospondin"
may refer to any one of the family of proteins which includes thrombospondins
I, II, III, IV, and
cartilage oligomeric matrix protein. Reference may also be made to one or more
of the specific
thrombospondins. Thrombospondin is a homotrimeric (TSP 1 and TSP2) or
homopentameric
(TSPs 3-5) glycoprotein with disulfide-linked subunits of MW 180,000. It
contains binding sites
for thrombin, fibrinogen, heparin, fibronectin, plasminogen, plasminogen
activator, collagen,
laminin, etc. It functions in many cell adhesion and migration events,
including platelet
aggregation.
[0125] Thrombospondin I(THBS1; also known as TSP1) has the Genbank accession
number X04665 for the human DNA sequence and TSP 1 human (P07996) for the
human protein
29


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
(see worldwide web at expasy.org/uniprot/P07996). It is a multimodular
secreted protein that
associates with the extracellular matrix and possesses a variety of biologic
functions, including a
potent angiogenic activity. Other thrombospondin genes include thrombospondins
II (THBS2;
188061), III (THBS3; 188062), and IV (THBS4; 600715) with the corresponding
protein
sequences TSP1 Human (P07996), TSP2 Human (P35442); TSP3 Human (P49746), and
TSP4
Human (P35443).
[0126] Human thrombospondin 2 (THBS2; also known as TSP2) has the Genbank
accession number L12350 (see worldwide web at expasy.org/uniprot/P35442) for
the human
sequence. It is very similar in sequence to THBS1.
[0127] Human thrombospondin 3 (THBS3; also known as TSP3) has the Genbank
accession number L38969 for the human sequence (see worldwide web at
expasy.org/uniprot/P49476). The protein is clearly homologous to THBSI and
THBS2 in its
COOH-terminal domains but substantially different in its NH2-terminal region,
suggesting
functional properties for THBS3 that are unique, but also related to those of
THBS 1 and THBS2.
The 956-amino acid predicted protein is highly acidic, especially in the third
quarter of the
sequence which corresponds to 7 type III calcium binding repeats. Four type II
EGF-like repeats
are also present.
[0128] The human THBS4 gene (also known as TSP4), Genbank accession number
Z19585
for the human sequence (see worldwide web at expasy.org/uniprot/P35443),
contains an RGD
(arg-gly-asp) cell-binding sequence in the third type 3 repeat. It is a
pentameric protein that binds
to heparin and calcium.
[0129] Cartilage oligomeric matrix protein (also known as TSP5), Genbank
accession
L32137 (see worldwide web at expasy.org/uniprot/P49474), is a 524-kD protein
that is expressed
at high levels in the territorial matrix of chondrocytes. The sequences
indicate that it is a member
of the thrombospondin gene family.

Thrombospondin EGF-like domains
[0130] In some embodiments of any of the methods described herein, the method
comprises
administering at least one thrombospondin EGF-like domain. The known
thrombospondin
isoforms comprise a number of specific domains. For THBSI and THBS2, these
include a
heparin-binding N terminal domain, a linker with homology to procollagen,
three TSP-type-1



CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
repeats, three EGF-like repeats, seven TSP type-3 calcium binding repeats, and
a cell-binding
carboxyl-terminal domain. As shown herein, for example in Figure 2, the EGF-
like domains are
sufficient to induce synaptogenesis.
[0131] The EGF-like domains have a distinct motif sequence. A common feature
is that
these repeats are found in the extracellular domain of membrane-bound proteins
or in proteins
known to be secreted. The EGF domain includes six cysteine residues which have
been shown to
be involved in disulphide bonds. The main structure is a two-stranded beta-
sheet followed by a
loop to a C-terminal short two-stranded sheet. Subdomains between the
conserved cysteines vary
in length. The disulfide bonded structure exemplified by EGF and its precursor
and TGF-a are
encountered in many proteins, e.g. as described by Apella et al. (1988) FEBS
Lett 231:1-4; and
Engel (1989) FEBS Lett 25:1-7, each herein incorporated by reference. An
alignment of the
mouse TSP EGF-like domains may be found in Bornstein (1992) FASEB J 6:3290-
3299, herein
incorporated by reference. Each EGF-like domain is from about 35 to about 70
amino acids in
length, more usually from around about 40 amino acids to around about 65 amino
acids in
length. The EGF repeat may include the hydroxylated amino acids, 0-
hydroxyaspartic acid and
(3-hydroxyasparagine. The repeats may also comprise negatively charged amino
acids at
positions 2, 4 and 5 of the domain.
[0132] Exemplary EGF-like domains are found in the human thrombospondin
polypeptides.
With reference to THBSI sequence and the above referenced sequence, EGF-like
domains are
found at amino acids 551-586, 588-636 and 650-689. With reference to THBS2
sequence and the
above referenced sequence, EGF-like domains are found at amino acids 553-588,
590-635, and
652-691. With reference to THBS3 sequence and the above referenced sequence,
EGF-like
domains are found at amino acids 316-368, 370-412, 418-455. With reference to
THBS4
sequence and the above referenced sequence, EGF-like domains are found at
amino acids 290-
324, 326-377, 379-418 and 424-461.
[0133] As used herein, a thrombospondin EGF-like domain includes, without
limitation, a
polypeptide of from about 35 to about 65 amino acids in length, comprising at
least 6 cysteine
amino acids, where the main structure is a two-stranded beta-sheet followed by
a loop to a C-
terminal short two-stranded sheet. The domain may have at least about 95%
sequence identity, at
least about 98% sequence identity, at least about 99% sequence identity, 100%
sequence identity

31


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
to the above defined domains from human thrombospondin proteins. Peptides may
be truncated
by 1, 2, 3, 4, 5, or more amino acids from the amino terminus, the carboxy
terminus, or both.
Agonists
[0134] The present invention provides methods for modulating axonal growth
and/or
synaptogenesis in an individual in need thereof comprising administering an
effective dose of an
agonist of a thrombospondin. In some embodiments, the invention provides
methods for
promoting synaptogenesis in an individual comprising administering to the
individual in need of
synaptogenesis an effective dose of an agonist. In some embodiments, the
agonist is a
thrombospondin agonist. Agonists may be tested for one or more agonist
activity (e.g., binds
and activates an a28 subunit of a calcium channel) using methods known in the
art and/or
methods described herein.
[0135] In some embodiments of any of the methods, thrombospondin agonists
include
polypeptides, nucleic acids, carbohydrates, immunoadhesion, thrombospondin
variants,
peptidomimetics, and small molecules, anti-thrombospondin antibodies and
immunoglobulin
variants, amino acid variants of human thrombospondin including amino acid
substitution,
deletion, and addition variants, or any combination thereof, and chimeric
immunoglobulins. The
thrombospondin agonists of this invention may be based on the inventors'
identification of the
thrombospondin domains involved in the binding of thrombospondin to its native
ligands, such
as a calcium channel a28 subunit. In some embodiments, thrombospondin agonists
are
antibodies. In some embodiments, the antibodies are monoclonal antibodies. In
some
embodiments, the monoclonal antibodies are chimeric antibodies or humanized
antibodies. In
some embodiments, the thrombospondin agonists are small molecules.
[0136] In some embodiments, thrombospondin agonists are polypeptides. In some
embodiments, the polypeptide is a polypeptide comprising at least one
thrombospondin EGF-like
domain, wherein the polypeptide binds and activates a calcium channel subunit
selected from the
group consisting of a251, a282, a263, and a254, and wherein synapse formation
in the individual
is increased. In some embodiments, the polypeptide is not a thrombospondin. In
some
embodiments, the polypeptide is a thrombospondin.
[0137] In some embodiments, the polypeptide thrombospondin agonist includes an
EGF-
like domain. EGF-like domain polypeptides of interest may include 1, 2, 3 or
more

32


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
thrombospondin EGF-like domains, as defined above. In some embodiments, the
EGF-like
domain polypeptides may comprise or consist of one EGF-like domain. In some
embodiments,
the EGF-like domain polypeptides include at least two thrombospondin EGF-like
domains. In
some embodiments, the EGF-like domain polypeptides include at least three
thrombospondin
EGF-like domains. In some embodiments, the EGF-like domain polypeptides
comprise the third
EGF-like domain, calcium binding repeats and C-terminal region of a
thrombospondin. In some
embodiments, the EGF-like domain polypeptides lack thrombospondin sequences
other than the
EGF-like domains. In some embodiments, the EGF-like domain polypeptide may
lack one or
more of the thrombospondin laminin G domain, von Willebrand factor C type
domain,
thrombospondin type I domain, thrombospondin type 3 repeat, and/or
thrombospondin C-
terminal region. In some embodiments, the thrombospondin EGF-like domain is a
polypeptide
derived from a thrombospondin isotype of from about 35 to about 65 amino acids
in length,
comprising at least 6 cysteine amino acids, where the main structure is a two-
stranded beta-sheet
followed by a loop to a C-terminal short two-stranded sheet. In some
embodiments, the
thrombospondin EGF-like domain has at least 95% sequence identity to human
THBS 1, amino
acids 551-586, 588-636, 650-689, human THBS2 amino acids 553-588, 590-635, 652-
691,
human THBS3 amino acids 316-368, 370-412, 418-455, human THBS4 amino acids 290-
324,
326-377, 379-418 and 424-461.
[0138] The sequence of the thrombospondin EGF-like domains may be altered in
various
ways known in the art to generate targeted changes in sequence. The
polypeptide will usually be
substantially similar to the sequences provided herein, i.e. will differ by at
least one amino acid,
and may differ by at least two but not more than about ten amino acids. The
sequence changes
may be substitutions, insertions or deletions. Scanning mutations that
systematically introduce
alanine, or other residues, may be used to determine key amino acids.
Conservative amino acid
substitutions typically include substitutions within the following groups:
(glycine, alanine);
(valine, isoleucine, leucine); (aspartic acid, glutamic acid); (asparagine,
glutamine); (serine,
threonine); (lysine, arginine); or (phenylalanine, tyrosine).
[0139] Modifications of interest that do not alter primary sequence include
chemical
derivatization of polypeptides, e.g., acetylation, or carboxylation. Also
included are
modifications of glycosylation, e.g. those made by modifying the glycosylation
patterns of a
polypeptide during its synthesis and processing or in further
processing.steps; e.g. by exposing

33


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
the polypeptide to enzymes which affect glycosylation, such as mammalian
glycosylating or
deglycosylating enzymes. Also embraced are sequences that have phosphorylated
amino acid
residues, e.g. phosphotyrosine, phosphoserine, or phosphothreonine.
[0140] Also included in the subject invention are polypeptides that have been
modified
using ordinary molecular biological techniques and synthetic chemistry so as
to improve their
resistance to proteolytic degradation or to optimize solubility properties or
to render them more
suitable as a therapeutic agent. For examples, the backbone of the peptide may
be cyclized to
enhance stability (see Friedler et al. (2000) J. Biol. Chem. 275:23783-23789).
Analogs of such
polypeptides include those containing residues other than naturally occurring
L-amino acids, e.g.
D-amino acids or non-naturally occurring synthetic amino acids. In some
embodiments,
thrombospondin polypeptides, including their immunogenic epitopes and other
fragments, may
be combined with heterologous molecules, resulting in therapeutically useful
fusion molecules. It
provides fusion partners capable of imparting favorable pharmacokinetics
and/or
pharmacodynamics to the thrombospondin. In an embodiment, the invention
provides a fusion
molecule comprising all or a part of the EGF-like domains of thrombospondin,
or fragments
thereof and a fusion partner, such as an antibody Fc domain. Fusion molecules
of the invention
may have an increased half-life in vivo, as compared to thrombospondin EGF-
like domains.
[0141] The subject peptides may be prepared by in vitro synthesis, using
conventional
methods as known in the art. Various commercial synthetic apparatuses are
available, for
example, automated synthesizers by Applied Biosystems, Inc., Foster City, CA,
Beckman, etc.
By using synthesizers, naturally occurring amino acids may be substituted with
unnatural amino
acids. The particular sequence and the manner of preparation will be
determined by convenience,
economics, purity required, and the like.
[0142] If desired, various groups may be introduced into the peptide during
synthesis or
during expression, which allow for linking to other molecules or to a surface.
Thus cysteines can
be used to make thioethers, histidines for linking to a metal ion complex,
carboxyl groups for
forming amides or esters, amino groups for forming amides, and the like.
[0143] The polypeptides may also be isolated and purified in accordance with
conventional
methods of recombinant synthesis. A lysate may be prepared of the expression
host and the
lysate purified using HPLC, exclusion chromatography, gel electrophoresis,
affinity
chromatography, or other purification technique. For the most part, the
compositions which are

34


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
used will comprise at least 20% by weight of the desired product, more usually
at least about
75% by weight, preferably at least about 95% by weight, and for therapeutic
purposes, usually at
least about 99.5% by weight, in relation to contaminants related to the method
of preparation of
the product and its purification. Usually, the percentages will be based upon
total protein.
Antagonists
[0144] The present invention provides methods for modulating axonal growth
and/or
synaptogenesis in an individual in need thereof comprising administering an
effective dose of an
antagonist (e.g., agent) of a thrombospondin and/or a calcium channel. In some
embodiments,
the methods inhibit synaptogenesis, decrease the number of synapses, or
decrease the activity of
synapses. In some embodiments, the methods promote axonal growth. In some
embodiments, the
methods promote dendritic growth. In some embodiments, the methods promote
both axonal and
dendritic growth. In some embodiments, the antagonist is a thrombospondin
antagonist. In some
embodiments, the thrombospondin antagonist inhibits one or more activities of
a
thrombospondin, for example, synaptogenesis activity. In some embodiments, the
thrombospondin antagonist binds to a thrombospondin and blocks the interaction
between a
thrombospondin and an a26 subunit of a calcium channel. In some embodiments,
the antagonist
is an antagonist to an a26 subunit of a calcium channel. In some embodiments,
the antagonist
binds to an a26 subunit of a calcium channel and blocks the interaction
between a
thrombospondin and the a28 subunit of a calcium channel.
[0145] Further, the invention provides, in some embodiments, methods for
treating pain
(such as neuropathic pain, visceral pain, cancer pain, inflammatory pain, post
operative pain,
migraine pain, or phantom pain) in an individual comprising administering to
an individual
having pain an effective amount of an antagonist. In some embodiments, the
pain includes, but is
not limited to, somatic pain (e.g. cutaneous (body surface) or deep tissues
(musculoskeletal
tissues) pain); pain associated with ligaments, tendons, bones, blood vessels,
fasciae, and
muscles; visceral pain (e.g. thoracic (chest) pain, abdominal pain, or pelvic
viscera pain, or
neuropathic pain); any pain caused by injury to the nervous system,
chemotherapy, radiation,
surgery, tumor compressing, or accident; cancer pain (e.g., breakthrough
cancer pain, pain from
pancreatic cancer, or metastases in the abdomen or bone); inflammatory pain
(e.g. pain
associated with rheumatoid arthritis, psoriatic arthritis, Reiter's syndrome
(reactive arthritis),



CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
ankylosing spondylitis, or inflammatory arthritic disorders); post operative
pain; migraine pain;
or phantom pain (e.g. amputation in quadriplegics). In some embodiments, the
pain can be either
acute or chronic, and comprises allodynia (i.e. pain due to a stimulus which
does not normally
provoke pain) or hyperalgesia (i.e. an increased response to a stimulus which
is normally
painful).
[0146] In some embodiments, the invention also provides methods for treating
epilepsy in
an individual comprising administering to the individual an effective amount
of an antagonist. In
some embodiments, the antagonist is a thrombospondin antagonist. In some
embodiments, the
antagonist is a a28 subunit calcium channel antagonist. In some embodiments,
the methods
comprise administering to an individual in need thereof an effective amount of
an antagonist
(e.g., agent) that binds to a thrombospondin and blocks the interaction
between the
thrombospondin and one or more calcium channel subunits selected from the
group consisting of
a261, a282, a283, and a264.
[0147] Antagonists of synaptogenesis include agents that interfere with the
interaction
between thrombospondin and calcium channel subunit a28, which include, without
limitation,
antibodies specific for thrombospondin, particularly antibodies specific for
at least one
thrombospondin EGF-like domain, and a26 ligands, including gabapentin and
pregabalin and
analogs thereof, e.g. as described in US Patent nos. 6518289, 6683112,
4087544, published US
Patent application 20050192353, etc., herein incorporated by reference. In
some embodiments,
the antagonists exclude gabapentin, pregabalin and/or analogs thereof.
[0148] In some embodiments of any of the methods, thrombospondin antagonists
include
polypeptides, nucleic acids (e.g., RNAs (e.g., siRNA, antisense RNA, or
microRNA), and
DNAs), carbohydrates, immunoadhesion, thrombospondin variants, thrombospondin
peptide
antagonists, peptidomimetics, small molecules, anti- thrombospondin antibodies
and
immunoglobulin variants, scaffold-derived binding proteins that binds to a
thrombospondin.
Scaffold-based proteins comprise single domains of antibodies, the
immunoglobulin
superfamily, protease inhibitors, helix-bundle proteins, disulphide-knotted
peptides, protein A,
lipocalins, fibronectin domains, ankyrin consensus repeat domains,
thioredoxins, and high
disulfide density scaffold proteins. See e.g., U.S. 6818418, Lipovsek et al.;
Skerra (2000) JMoI
Recognit. 13(4):167-87; Skerra (2007) Current Opinion in Biotechnology, 18:
295-304;
worldwide web at.amunix.com/Technology.html. Amino acid variants of human
thrombospondin

36


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
include amino acid substitution, deletion, and addition variants, or any
combination thereof, and
chimeric immunoglobulins. The thrombospondin antagonists of this invention may
be based on
the inventors' identification of the thrombospondin domains involved in the
binding of
thrombospondin to a calcium channel a26 subunit. In some embodiments,
thrombospondin
antagonists are antibodies. In some embodiments, the antibodies are monoclonal
antibodies. In
some embodiments, the monoclonal antibodies are chimeric antibodies or
humanized antibodies.
In some embodiments, the thrombospondin antagonists are small molecules. In
some
embodiments, the small molecule is gabapentin or an analog thereof. In some
embodiments, the
small molecule is a small molecule other than gabapentin or an analog thereof.
[0149] In some embodiments, the antagonist (e.g., agent) specifically binds to
a
thrombospondin. In some embodiments, the antagonist specifically binds to an
EGF-like domain
of the thrombospondin. In some embodiments, the antagonist specifically binds
to the third EGF-
like domain of the thrombospondin. In some embodiments, thrombospondin is TSP
1, TSP2,
TSP3, TSP4, or cartilage oligomeric matrix. In some embodiments, the
antagonist is an antibody.
[0150] In some embodiments, the antagonist is a protein scaffold for antibody
mimics or
scaffold-derived binding proteins that display properties like small size,
stability, and ease of
production. These include single domains of antibodies or the immunoglobulin
superfamily,
protease inhibitors, helix-bundle proteins, disulphide-knotted peptides,
protein A, the lipocalins,
fibronectin domains, ankyrin consensus repeat domains, thioredoxin, and high
disulfide density
scaffold proteins.
[0151] In some embodiments, the antagonist is a siRNA, an antisense RNA, or a
microRNA
that specifically inhibits expression of one or more thrombospondins. In some
embodiments, the
expression of TSP1, TSP2, TSP4, or cartilage oligomeric matrix is inhibited.
[0152] In some embodiments of any of the methods described herein, antagonists
of the a28
subunit of a calcium channel include polypeptides, nucleic acids (e.g., RNAs
(e.g., siRNA,
antisense RNA, or microRNA), and DNAs), carbohydrates, immunoadhesion, calcium
channel
variants, calcium channel peptide antagonists, peptidomimetics, and small
molecules, anti-
calcium channel antibodies and immunoglobulin variants, scaffold-derived
binding proteins that
bind to an a28 subunit (such as VWFA domain), amino acid variants of human
calcium channel
including amino acid substitution, deletion, and addition variants, or any
combination thereof,
and chimeric immunoglobulins. The antagonists of the a28 subunit of a calcium
channel of this

37


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
invention may be based on the inventors' identification of the calcium channel
domains involved
in the binding of calcium channel to thrombospondin. In some embodiments, the
antagonists of
the a25 subunit of a calcium channel are antibodies. In some embodiments, the
antibodies are
monoclonal antibodies. In some embodiments, the monoclonal antibodies are
chimeric
antibodies or humanized antibodies. In some embodiments, the antibodies are
antibodies to a28 1.
In some embodiments, the antibodies specifically bind to the VWFA domain of an
a281. In
some embodiments, the antagonists of the a28 subunit of a calcium channel are
small molecules.
In some embodiments, the small molecule is gabapentin or an analog thereof. In
some
embodiments, the small molecule is a small molecule other than gabapentin or
an analog thereof.
In some embodiments, a modulating and/or antagonist protein comprises the
polypeptide
sequence of S portion of the calcium channel subunit a28 (e.g., the 81 portion
of the calcium
channel subunit a28 1).
[0153] In some embodiments of any of the methods described herein, antagonists
include a
polypeptide comprising an extracellular portion of a calcium channel a28
subunit. In some
embodiments, polypeptide comprises the a2 portion of the calcium channel
subunit a28 subunit.
In some embodiments, polypeptide comprises the amino acids of about 253 to
about 430 of
human a281 (VWFA domain). In some embodiments, polypeptide comprises an
extracellular
portion of a calcium channel a282. In some embodiments, polypeptide comprises
the a2 portion
of the calcium channel subunit a262. In some embodiments, polypeptide
comprises the amino
acids of about'291 to about 469 of human a282 (VWFA domain). In some
embodiments,
polypeptide comprises an extracellular portion of a calcium channel a283. In
some embodiments,
polypeptide comprises the a2 portion of a calcium channel subunit a283. In
some embodiments,
polypeptide comprises the amino acids of about 256 to about 438 of human a283
(VWFA
domain). In some embodiments, polypeptide comprises an extracellular portion
of a calcium
channel a284. In some embodiments, polypeptide comprises the a2 portion of the
calcium
channel subunit a284. In some embodiments, polypeptide comprises the amino
acids of about
291 to about 472 of human a284. In some embodiments, polypeptide comprises the
amino acids
of about 291 to about 473 of human a284. (VWFA domain).
[0154] Polypeptide sequences (including the calcium channel subunit a25 such
as the
extracellular portion of the calcium channel subunit a28) may be altered in
various ways known
in the art to generate targeted changes in sequence. The polypeptide will
usually be substantially
38


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
similar to the sequences provided herein, i.e. will differ by at least one
amino acid, and may
differ by at least two but not more than about ten amino acids. The sequence
changes may be
substitutions, insertions or deletions. Scanning mutations that systematically
introduce alanine,
or other residues, may be used to determine key amino acids. Conservative
amino acid
substitutions typically include substitutions within the following groups:
(glycine, alanine);
(valine, isoleucine, leucine); (aspartic acid, glutamic acid); (asparagine,
glutamine); (serine,
threonine); (lysine, arginine); or (phenylalanine, tyrosine).
[0155] Modifications of interest that do not alter primary sequence include
chemical
derivatization of polypeptides, e.g., acetylation, or carboxylation. Also
included are
modifications of glycosylation, e.g. those made by modifying the glycosylation
patterns of a
polypeptide during its synthesis and processing or in further processing
steps; e.g. by exposing
the polypeptide to enzymes which affect glycosylation, such as mammalian
glycosylating or
deglycosylating enzymes. Also embraced are sequences that have phosphorylated
amino acid
residues, e.g. phosphotyrosine, phosphoserine, or phosphothreonine.
[0156] Also included in the subject invention are polypeptides that have been
modified
using ordinary molecular biological techniques and synthetic chemistry so as
to improve their
resistance to proteolytic degradation or to optimize solubility properties or
to render them more
suitable as a therapeutic agent. For examples, the backbone of the peptide may
be cyclized to
enhance stability (see Friedler et al. (2000) J. Biol. Chem. 275:23783-23789).
Analogs of such
polypeptides include those containing residues other than naturally occurring
L-amino acids, e.g.
D-amino acids or non-naturally occurring synthetic amino acids. In some
embodiments, the
calcium channel subunit a28 (e.g., the extracellular portion of the calcium
channel subunit a28),
including their immunogenic epitopes and other fragments, may be combined with
heterologous
molecules, resulting in therapeutically useful fusion molecules. It provides
fusion partners
capable of imparting favorable pharmacokinetics and/or pharmacodynamics to the
calcium
channel subunit a28 (e.g., the extracellular portion of the calcium channel
subunit a25). In an
embodiment, the invention provides a fusion molecule comprising all or a part
of the calcium
channel subunit a28 (e.g., the extracellular portion of the calcium channel
subunit a26), or
fragments thereof and a fusion partner, such as an antibody Fc domain. The Fc
domain may be
further modified to remove one or more effector functions. Fusion molecules of
the invention

39


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
may have an increased half-life in vivo, as compared to the calcium channel
subunit a28 (e.g., the
extracellular portion of the calcium channel subunit a26).
[0157] The subject peptides may be prepared by in vitro synthesis, using
conventional
methods as known in the art. Various commercial synthetic apparatuses are
available, for
example, automated synthesizers by Applied Biosystems, Inc., Foster City, CA,
Beckman, etc.
By using synthesizers, naturally occurring amino acids may be substituted with
unnatural amino
acids. The particular sequence and the manner of preparation will be
determined by convenience,
economics, purity required, and the like.
[0158] If desired, various groups may be introduced into the peptide during
synthesis or
during expression, which allow for linking to other molecules or to a surface.
Thus cysteines can
be used to make thioethers, histidines for linking to a metal ion complex,
carboxyl groups for
forming amides or esters, amino groups for forming amides, and the like.
[0159] The polypeptides may also be isolated and purified in accordance with
conventional
methods of recombinant synthesis. A lysate may be prepared of the expression
host and the
lysate purified using HPLC, exclusion chromatography, gel electrophoresis,
affinity
chromatography, or other purification technique. For the most part, the
compositions which are
used will comprise at least 20% by weight of the desired product, more usually
at least about
75% by weight, preferably at least about 95% by weight, and for therapeutic
purposes, usually at
least about 99.5% by weight, in relation to contaminants related to the method
of preparation of
the product and its purification. Usually, the percentages will be based upon
total protein.
Synapto-aenesis
[0160] Synaptogenesis is a dynamic process. During development, more synapses
are
established than ultimately will be retained. Therefore, the elimination of
excess synaptic inputs
is a critical step in synaptic circuit maturation. Synapse elimination is a
competitive process that
involves interactions between pre- and postsynaptic partners. In the CNS, as
with the NMJ, a
developmental, activity-dependent remodeling of synaptic circuits takes place
by a process that
may involve the selective stabilization of coactive inputs and the elimination
of inputs with
uncorrelated activity. The anatomical refinement of synaptic circuits occurs
at the level of
individual axons and dendrites by a dynamic process that involves rapid
elimination of synapses.
As axons branch and remodel, synapses form and dismantle with synapse
elimination occurring
rapidly.



CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
[0161] Synapses are asymmetric communication junctions formed between two
neurons, or,
at the neuromuscular junction (NMJ) between a neuron and a muscle cell.
Chemical synapses
enable cell-to-cell communication via secretion of neurotransmitters, whereas
in electrical
synapses signals are transmitted through gap junctions, specialized
intercellular channels that
permit ionic current flow. In addition to ions, other molecules that modulate
synaptic function
(such as ATP and second messenger molecules) can diffuse through gap
junctional pores. At the
mature NMJ, pre- and postsynaptic membranes are separated by a synaptic cleft
containing
extracellular proteins that form the basal lamina. Synaptic vesicles are
clustered at the
presynaptic release site, transmitter receptors are clustered in junctional
folds at the postsynaptic
membrane, and glial processes surround the nerve terminal.
[0162] In some embodiments of any of the methods described herein, synapse
formation
may be increased. In some embodiments, synapses are increased due to increased
new synapse
formation. In some embodiments, synapses are increased due to increased
synapse maintenance.
In some embodiments, the synapses are at the neuromuscular junction. In some
embodiments,
the synapses comprise or consist of excitatory synapses. In some embodiments,
the synapses are
VGlut2 positive excitatory synapses. In some embodiments, the synapses are
VGlutl positive
excitatory synapses. In some embodiments, the synapse formation is increased
after synapse loss
due to senescence. In some embodiments, the synapse formation is increase
after synapses loss
due to injury.
[0163] A number of cell adhesion molecules and tyrosine kinase receptor
ligands have been
implicated in modulating synaptogenesis. Integrins, cadherins, and
neuroligins, are cell adhesion
molecules that may play a role in synapse formation. The ephrins and their
receptors, the Eph
tyrosine kinases, participate in the activity-independent topographic
organization of brain circuits
and may also participate in synapse formation and maturation. Neurotrophins
have also been
implicated in aspects of synapse development and function. The methods of the
invention are
used to promote an improved outcome from ischemic cerebral injury, or other
neuronal injury,
by inducing synaptogenesis and cellular changes that promote functional
improvement. The
methods are also used to enhance synaptogenesis in patients suffering from
neurodegenerative
disorders, e.g. Alzheimer's disease, epilepsy, etc.

41


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
Diseases and Conditions of Interest
[0164] The methods described herein may be used to treat or prevent a variety
of diseases
and conditions. Among the conditions of interest for the present methods of
enhancing
synaptogenesis include, but not limited to, senescence, stroke, spinal cord
injury, Alzheimer's
disease (a disease where synapses are lost), Parkinson/s disease, multiple
sclerosis, amyotrophic
lateral sclerosis, neuropathy, mascular dystrophy, Huntington disease,
alcoholism , Alexander's
disease, Alper's disease ataxia telangiectasia, Batten disease (also known as
Spielmeyer-Vogt-
Sjogren-Batten disease), bovine spongiform encephalopathy (BSE), canavan
disease, Cockayne
syndrome, corticobasal degeneration , Creutzfeldt-Jakob disease, HIV-
associated dementia,
Kennedy's disease, Krabbe's disease, Lewy body dementia, Machado-Joseph
disease
(Spinocerebellar ataxia type 3) , multiple system atrophy, narcolepsy,
neuroborreliosis,
Pelizaeus-Merzbacher Disease, primary lateral sclerosis, prion diseases,
Progressive
Supranuclear Palsy, Refsum's disease, Sandhoff's disease, Schilder's disease,
subacute
combined degeneration of spinal cord secondary to pernicious anaemia,
schizophrenia,
Spielmeyer-Vogt-Sjogren-Batten disease (also known as Batten disease),
spinocerebellar ataxia
(multiple types with varying characteristics), spinal muscular atrophy, Steele-
Richardson-
Olszewski disease, Tabes dorsalis, as well as promoting new synaptogenesis in
repair and
regeneration of injured CNS after stroke or spinal cord injury. Such
conditions benefit from
administration of thrombospondin or thrombospondin agonists, which increase,
or enhance, the
development of synapses. In some instances, where there has been neuronal
loss, it may be
desirable to enhance neurogenesis as well, e.g. through administration of
agents or regimens that
increase neurogenesis, transplantation of neuronal progenitors, etc.
[0165] Patients can suffer neurological and functional deficits after stroke,
CNS injury, and
neurodegenerative disease. The findings of the present invention provide a
means to modulate
synapse formation and to improve function after CNS damage or degeneration.
The induction of
neural connections induced by promoting synaptogenesis will promote functional
improvement
after stroke, injury, aging and neurodegenerative disease. The amount of
increased
synaptogenesis may comprise at least a measurable increase relative to a
control lacking such
treatment, for example at least a 10% increase, at least a 20% increase, at
least a 50% increase, or
more. In some embodiments, the number of synapses may be increased at least
about any of
10%, 20%, 30%, 40%, 50%, or 60%. In some embodiments, the synapses are at the

42


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
neuromuscular junction. In some embodiments, the synapses comprise or consist
of excitatory
synapses. In some embodiments, the synapses are VGlut2 positive excitatory
synapses. In some
embodiments, the synapses are VGlutl positive excitatory synapses. In some
embodiments,
synapses are increased due to increased new synapse formation. In some
embodiments, synapses
are increased due to increased synapse maintenance.
[0166] In some embodiments of any of the methods described herein, an
individual or
subject may have suffered synapse loss as a result of senescence. In some
embodiments, the
individual or subject may have suffered synapse loss as a result of
Alzheimer's disease,
Parkinson's disease, ALS, multiple sclerosis, or glaucoma. In some
embodiments, an individual
or subject may have suffered macular degeneration, a hearing loss, diabetic
neuropathy, or
chemotherapy induced neuropathy. In some embodiments, the individual or
subject may have
suffered synapse loss as a result of a psychiatric disorder selected from the
group consisting of
acute stress disorder, agoraphobia, dissociative amnesia, anorexia nervosa,
bipolar disorder, body
dysmorphic disorder, brief psychotic disorder, bulimia nervosa, conversion
disorder,
cyclothymic disorder, delusional disorder, depersonalization disorder,
dissociative identity
disorder (DID), dysparenunia, dysthymic disorder, male erectile disorder,
generalized anxiety
disorder, impotence, pain disorder, panic disorder, phobias, posttraumatic
stress disorder,
schizoaffective disorder, schizophreniform, shared psychotic disorder, and
substance abuse. In
some embodiments, the individual may have suffered synapse loss due to injury
such as spinal
cord injury or central nervous system injury.
[0167] The term "stroke" broadly refers to the development of neurological
deficits
associated with impaired blood flow to the brain regardless of cause.
Potential causes include,
but are not limited to, thrombosis, hemorrhage and embolism. Current methods
for diagnosing
stroke include symptom evaluation, medical history, chest X-ray, ECG
(electrical heart activity),
EEG (brain nerve cell activity), CAT scan to assess brain damage and MRI to
obtain internal
body visuals. Thrombus, embolus, and systemic hypotension are among the most
common
causes of cerebral ischemic episodes. Other injuries may be caused by
hypertension,
hypertensive cerebral vascular disease, rupture of an aneurysm, an angioma,
blood dyscrasias,
cardiac failure, cardiac arrest, cardiogenic shock, septic shock, head trauma,
spinal cord trauma,
seizure, bleeding from a tumor, or other blood loss.

43


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
[0168] By "ischemic episode" is meant any circumstance that results in a
deficient supply
of blood to a tissue. When the ischemia is associated with a stroke, it can be
either global or focal
ischemia, as defined below. The term "ischemic stroke" refers more
specifically to a type of
stroke that is of limited extent and caused due to blockage of blood flow.
Cerebral ischemic
episodes result from a deficiency in the blood supply to the brain. The spinal
cord, which is also
a part of the central nervous system, is equally susceptible to ischemia
resulting from diminished
blood flow.
[0169] Senescence refers to the effects or the characteristics of increasing
age, particularly
with respect to the diminished ability of somatic tissues to regenerate in
response to damage,
disease, and normal use. Alternatively, aging may be defined in terms of
general physiological
characteristics. The rate of aging is very species specific, where a human may
be aged at about
50 years; and a rodent at about 2 years. In general terms, a natural
progressive decline in body
systems starts in early adulthood, but it becomes most evident several decades
later. One
arbitrary way to define old age more precisely in humans is to say that it
begins at conventional
retirement age, around about 60, around about 65 years of age. Another
definition sets
parameters for aging coincident with the loss of reproductive ability, which
is around about age
45, more usually around about 50 in humans, but will, however, vary with the
individual. Loss of
synaptic function may be found in aged individuals, such as mild cognitive
deficient..
[0170] Among the aged, Alzheimer's disease is a serious condition. Alzheimer's
disease is
a progressive, inexorable loss of cognitive function associated with an
excessive number of
senile plaques in the cerebral cortex and subcortical gray matter, which also
contains 0-amyloid
and neurofibrillary tangles consisting of tau protein. The common form affects
persons > 60 yr
old, and its incidence increases as age advances. It accounts for more than
65% of the dementias
in the elderly.
[0171] The cause of Alzheimer's disease is not known. The disease runs in
families in about
15 to 20% of cases. The remaining, so-called sporadic cases have some genetic
determinants.
The disease has an autosomal dominant genetic pattern in most early-onset and
some late-onset
cases but a variable late-life penetrance. Environmental factors are the focus
of active
investigation.
[0172] In the course of the disease, neurons are lost within the cerebral
cortex,
hippocampus, and subcortical structures (including selective cell loss in the
nucleus basalis of
44


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
Meynert), locus caeruleus, and nucleus raphae dorsalis. Cerebral glucose use
and perfusion is
reduced in some areas of the brain (parietal lobe and temporal cortices in
early-stage disease,
prefrontal cortex in late-stage disease). Neuritic or senile plaques (composed
of neurites,
astrocytes, and glial cells around an amyloid core) and neurofibrillary
tangles (composed of
paired helical filaments) play a role in the pathogenesis of Alzheimer's
disease. Senile plaques
and neurofibrillary tangles occur with normal aging, but they are much more
prevalent in persons
with Alzheimer's disease.
[0173] The essential features of dementia are impairment of short-term memory
and long-
term memory, abstract thinking, and judgment; other disturbances of higher
cortical function;
and personality change. Progression of cognitive impairment confirms the
diagnosis, and patients
with Alzheimer's disease do not improve.
[0174] The methods of the invention also find use in combination with cell or
tissue
transplantation to the central nervous system, where such grafts include
neural progenitors such
as those found in fetal tissues, neural stem cells, embryonic stem cells or
other cells and tissues
contemplated for neural repair or augmentation. Neural stem/progenitor cells
have been
described in the art, and their use in a variety of therapeutic protocols has
been widely discussed.
For example, inter alia, U.S. Patent nos. 6,638,501, Bjomson et al.; U.S.
6,541,255, Snyder et
al.; U.S. 6,498,018, Carpenter; U.S. Patent Application 20020012903, Goldman
et al.; Palmer et
al. (2001) Nature 411(6833):42-3; Palmer et al. (1997) Mol Cell Neurosci.
8(6):389-404;
Svendsen et al. (1997) Exp. Neurol. 148(1):135-46 and Shihabuddin (1999) Mol
Med Today
5(11):474-80; each herein specifically incorporated by reference.
[0175] Neural stem and progenitor cells can participate in aspects of normal
development,
including migration along well-established migratory pathways to disseminated
CNS regions,
differentiation into multiple developmentally- and regionally-appropriate cell
types in response
to microenvironmental cues, and non-disruptive, non-tumorigenic interspersion
with host
progenitors and their progeny. Human NSCs are capable of expressing foreign
transgenes in vivo
in these disseminated locations. As such, these cells find use in the
treatment of a variety of
conditions, including traumatic injury to the spinal cord, brain, and
peripheral nervous system;
treatment of degenerative disorders including Alzheimer's disease,
Huntington's disease,
Parkinson's disease; affective disorders including major depression; stroke;
and the like. By



CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
synaptogenesis enhancers, the functional connections of the neurons are
enhances, providing for
an improved clinical outcome.
101761 Where a synaptogenesis inhibitor of the invention is administered, the
decrease in
synaptogenesis may comprise at least a measurable decrease relative to a
control lacking such
treatment, for example at least a 10% decrease, at least a 20% decrease, at
least a 50% decrease,
or more. In some embodiments, the number of synapses may be inhibited at least
about any of
10%, 20%, 30%, 40%, 50%, or 60%. In some embodiments, the synapses are at the
neuromuscular junction. In some embodiments, the synapses comprise or consist
of excitatory
synapses. In some embodiments, the synapses are VG1ut2 positive excitatory
synapses. In some
embodiments, the synapses are VGlutl positive excitatory synapses.
[0177] Among the conditions of interest for the present methods of decreasing
synaptogenesis are in the treatment of pain, epilepsy, anxiety, addiction, and
to aid in the axon
growth of regenerating neurons. Such conditions benefit from administration of
thrombospondin
antagonists, which decrease, or inhibit, the development of synapses. Any
antagonists described
herein may be used, such as antibodies and fragments thereof that specifically
bind to a
thrombospondin (such as the EGF-like domain of a thrombospondin); and
molecules that bind to
a calcium channel a28 subunit (e.g., an a26 subunit of calcium channel, such
as a28 1, a282,
a283, and a254), e.g. an antibody that specifically binds to VWFA domain of an
a26, gapapentin
and analogs thereof, particularly including analogs identified by the
screening methods described
herein.
[0178] Epilepsy is a recurrent, paroxysmal disorder of cerebral function
characterized by
sudden, brief attacks of altered consciousness, motor activity, sensory
phenomena, or
inappropriate behavior caused by excessive discharge of cerebral neurons.
Manifestations
depend on the type of seizure, which may be classified as partial or
generalized. In partial
seizures, the excess neuronal discharge is contained within one region of the
cerebral cortex. In
generalized seizures, the discharge bilaterally and diffusely involves the
entire cortex.
Sometimes a focal lesion of one part of a hemisphere activates the entire
cerebrum bilaterally so
rapidly that it produces a generalized tonic-clonic seizure before a focal
sign appears.
[0179) Most patients with epilepsy become neurologically normal between
seizures,
although overuse of anticonvulsants can dull alertness. Progressive mental
deterioration is
usually related to the neurologic disease that caused the seizures. Left
temporal lobe epilepsy is

46


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
associated with verbal memory abnormalities; right temporal lobe epilepsy
sometimes causes
visual spatial memory abnormalities. The outlook is best when no brain lesion
is demonstrable.
[0180] If a peripheral nerve is partially damaged, function is restored before
the severed
fibers regenerate. The peripheral nervous system, spinal cord and brain have
all been shown to
sprouting and circuitry remodeling. Following injury, regions not primarily
associated with the
lesion also exhibit synaptic density changes and subsequent recovery of
control levels over a
long period of time. The synaptic changes occur despite the absence of
degenerating terminals
within these zones. Thus, pronounced transneuronal changes may occur after
major trauma to the
CNS, suggesting that reactive synaptogenesis may adjust the functional
integrity of complex
circuitry in areas with and without a primary lesion. When an injury occurs in
the mature brain,
the growth process must be executed in the context of a damaged system. The
old system must
be cleared and coordinated with the initiation of growth and the formation of
new synapses. The
capacity for extensive remodeling and growth is desirably restrained when such
remodeling is
not required. In such conditions, an inhibitor of synaptogenesis according to
the present
invention may be administered for a period of time sufficient to permit neuron
growth, prior to
synapse formation.
[0181] Synaptogenesis is involved in the underlying neural basis of alcoholism
and drug
addiction. Neurobiological studies have identified specific brain areas and
molecular
mechanisms involved in drug abuse and dependence. Drug-induced persistent
behaviors such as
sensitization, tolerance or relapse far outlast any previously reported
molecular mechanisms.
Ultrastructural evidence of synaptic rewiring has been found in association
with cocaine-induced
behavioral sensitization. Such synaptic remodeling represents a potential
neural substrate
underlying the persistence of addiction. Site-specific pharmacotherapeutic and
behavioral
treatment programs for alcoholism and drug addiction can then target these
circuits. In such
conditions, an inhibitor of synaptogenesis according to the present invention
may be
administered for a period of time sufficient to inhibit synapse formation
associated with
addiction.
[0182] The findings of the present invention also provide a means to modulate
axonal
and/or dendritic growth and to improve function after CNS damage. The
induction of axonal
and/or dendritic growth will promote functional improvement after injury. The
amount of
increase in axonal and/or dendritic growth may comprise at least a measurable
increase relative

47


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747

to a control lacking such treatment, for example at least a 10% increase, at
least a 20% increase,
at least a 50% increase, or more. In some embodiments, the axonal and/or
dendritic growth may
be increased at least about any of 10%, 20%, 30%, 40%, 50%, or 60%. In some
embodiments,
the axonal and/or dendritic growth is axon growth. In some embodiments, the
axonal and/or
dendritic growth is dendritic growth. Where an axonal and/or dendritic growth
inhibitor of the
invention is administered, the decrease in axonal and/or dendritic growth may
comprise at least a
measurable decrease relative to a control lacking such treatment, for example
at least a 10%
decrease, at least a 20% decrease, at least a 50% decrease, or more. In some
embodiments, the
axonal and/or dendritic growth may be inhibited at least about any of 10%,
20%, 30%, 40%,
50%, or 60%. In some embodiments, the axonal and/or dendritic growth inhibited
is axonal
growth. In some embodiments, the axonal and/or dendritic growth inhibited is
dendritic growth.
[0183] In some embodiments of any of the methods described herein, the
individual may
have suffered axonal and/or dendritic degeneration as a result of a spinal
cord injury. In some
embodiments, the individual has suffered axonal and/or dendritic degeneration
as a result of
Alzheimer's disease, Parkinson's disease, ALS, or multiple sclerosis. In some
embodiments, the
individual has suffered a macular degeneration, a hearing loss, a diabetic
neuropathy, or a
chemotherapy induced neuropathy. In some embodiments, the individual has
suffered axonal
and/or dendritic degeneration as a result of a psychiatric disorder selected
from the group
consisting of acute stress disorder, agoraphobia, dissociative amnesia,
anorexia nervosa, bipolar
disorder, body dysmorphic disorder, brief psychotic disorder, bulimia nervosa,
conversion
disorder, cyclothymic disorder, delusional disorder, depersonalization
disorder, dissociative
identity disorder (DID), dysparenunia, dysthymic disorder, male erectile
disorder, generalized
anxiety disorder, impotence, pain disorder, panic disorder, phobias,
posttraumatic stress disorder,
schizoaffective disorder, schizophreniform, shared psychotic disorder, and
substance abuse..
[0184] The antagonists described herein may also be used for treating or
preventing a
disorder characterized by excess of calcium influx in an individual. The
invention provides
methods comprising administering to the individual an effective amount of an
agent that blocks
the interaction between a thrombospondin and a calcium subunit selected from
the group
consisting of a251, a252, a283, and a284. In some embodiments, the agent (such
as an
antibody) specifically binds to a thrombospondin. In some embodiments, the
disorder is muscle
spasm, migraine, stroke, or Parkinson's disease.

48


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
Gene Delivery
[0185] One approach for modulating synaptogenesis involves gene therapy. In
such
methods, sequences encoding an agonist or an antagonist described herein, such
as peptide
comprising at least one thrombospondin EGF-like domain, RNAi sequence, or
antibodies, are
introduced into the central nervous system, and expressed, as a means of
providing agonist or
antagonist activity to the targeted cells. To genetically modify neurons that
are protected by the
BBB, two general categories of approaches have been used. In one type of
approach, cells are
genetically altered, outside the body, and then transplanted somewhere in the
CNS, usually in an
area inside the BBB. In the other type of approach, genetic "vectors" are
injected directly into
one or more regions in the CNS, to genetically alter cells that are normally
protected by the
BBB. It should be noted that the terms "transfect" and "transform" are used
interchangeably
herein. Both terms refer to a process which introduces a foreign gene (also
called an
"exogenous" gene) into one or more preexisting cells, in a manner which causes
the foreign
gene(s) to be expressed to form corresponding polypeptides.
[0186] A preferred approach aims to introduce into the CNS a source of a
desirable
polypeptide, by genetically engineering cells within the CNS. This has been
achieved by directly
injecting a genetic vector into the CNS, to introduce foreign genes into CNS
neurons "in situ"
(i.e., neurons which remain in their normal position, inside a patient's brain
or spinal cord,
throughout the entire genetic transfection or transformation procedure).
[0187] Useful vectors include viral vectors, which make use of the lipid
envelope or surface
shell (also known as the capsid) of a virus. These vectors emulate and use a
virus's natural ability
to (i) bind to one or more particular surface proteins on certain types of
cells, and then (ii) inject
the virus's DNA or RNA into the cell. In this manner, viral vectors can
deliver and transport a
genetically engineered strand of DNA or RNA through the outer membranes of
target cells, and
into the cells cytoplasm. Gene transfers into CNS neurons have been reported
using such vectors
derived from herpes simplex viruses (e.g., European Patent 453242, Breakfield
et al 1996),
adenoviruses (La Salle et al 1993), and adeno-associated viruses (Kaplitt et
al 1997).
[0188] Non-viral vectors typically contain the transcriptional regulatory
elements necessary
for expression of the desired gene, and may include an origin of replication,
selectable markers
and the like, as known in the art. The non-viral genetic vector is then
created by adding, to a gene

49


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
expression construct, selected agents that can aid entry of the gene construct
into target cells.
Several commonly-used agents include cationic lipids, positively charged
molecules such as
polylysine or polyethylenimine, and/or ligands that bind to receptors
expressed on the surface of
the target cell. For the purpose of this discussion, the DNA-adenovirus
conjugates described by
Curiel (1997) are regarded as non-viral vectors, because the adenovirus capsid
protein is added
to the gene expression construct to aid the efficient entry of the gene
expression construct into
the target cell.
[0189] In cationic gene vectors, DNA strands are negatively charged, and cell
surfaces are
also negatively charged. Therefore, a positively-charged agent can help draw
them together, and
facilitate the entry of the DNA into a target cell. Examples of positively-
charged transfection
agents include polylysine, polyethylenimine (PEI), and various cationic
lipids. The basic
procedures for preparing genetic vectors using cationic agents are similar. A
solution of the
cationic agent (polylysine, PEI, or a cationic lipid preparation) is added to
an aqueous solution
containing DNA (negatively charged) in an appropriate ratio. The positive and
negatively
charged components will attract each other, associate, condense, and form
molecular complexes.
If prepared in the appropriate ratio, the resulting complexes will have some
positive charge,
which will aid attachment and entry into the negatively charged surface of the
target cell. The
use of liposomes to deliver foreign genes into sensory neurons is described in
various articles
such as Sahenk et al 1993. The use of PEI, polylysine, and other cationic
agents is described in
articles such as Li et al 2000 and Nabel et al 1997.
[0190] An alternative strategy for introducing DNA into target cells is to
associate the DNA
with a molecule that normally enters the cell. This approach was demonstrated
in liver cells in
U.S. Pat. No. 5,166,320 (Wu et al 1992). An advantage of this approach is that
DNA delivery
can be targeted to a particular type of cell, by associating the DNA with a
molecule that is
selectively taken up by that type of target cell. A limited number of
molecules are known to
undergo receptor mediated endocytosis in neurons. Known agents that bind to
neuronal receptors
and trigger endocytosis, causing them to enter the neurons, include (i) the
non-toxic fragment C
of tetanus toxin (e.g., Knight et al 1999); (ii) various lectins derived from
plants, such as barley
lectin (Horowitz et al 1999) and wheat germ agglutinin lectin (Yoshihara et al
1999); and, (iii)
certain neurotrophic factors (e.g., Barde et al 1991). At least some of these
endocytotic agents
undergo "retrograde" axonal transport within neuron. The term "retrograde", in
this context,



CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
means that these molecules are actively transported, by cellular processes,
from the extremities
(or "terminals") of a neuron, along an axon or dendrite, toward and into the
main body of the
cell, where the nucleus is located. This direction of movement is called
"retrograde", because it
runs in the opposite direction of the normal outward ("anterograde") movement
of most
metabolites inside the cell (including proteins synthesized in the cell body,
neurotransmitters
synthesized by those proteins, etc.).
Methods ofAdministration and Dosages
[0191] As such, administration of the compounds (including agonists and
antagonists
described herein) can be achieved in various ways, including oral, buccal,
rectal, parenteral,
intraperitoneal, intradermal, transdermal, intrathecal, nasal, intranasal,
topical, intravenous,
intraarterial, intramuscular, subcutaneous, subdermal, intracranial,
ophthalmic (e.g., topical,
injection (e.g., subconjunctival, subtenon, intravitreal, etc.), or
implantation), or intrathecal
administration. The active agent may be systemic after administration or may
be localized by the
use of regional administration, intramural administration, or use of an
implant that acts to retain
the active dose at the site of implantation.
[0192] The compositions of the invention may be administered using any
medically
appropriate procedure, e.g. intravascular (intravenous, intraarterial,
intracapillary)
administration, injection into the cerebrospinal fluid, intracavity or direct
injection in the brain.
Intrathecal administration maybe carried out through the use of an Ommaya
reservoir, in
accordance with known techniques. (F. Balis et al., Am J. Pediatr. Hematol.
Oncol. 11, 74, 76
(1989).
[0193] One strategy for drug delivery through the blood brain barrier (BBB)
entails
disruption of the BBB, either by osmotic means such as mannitol or
leukotrienes, or
biochemically by the use of vasoactive substances such as bradykinin. The
potential for using
BBB opening to target specific agents is also an option. A BBB disrupting
agent can be co-
administered with the therapeutic compositions of the invention when the
compositions are
administered by intravascular injection. Other strategies to go through the
BBB may entail the
use of endogenous transport systems, including carrier-mediated transporters
such as glucose and
amino acid carriers, receptor-mediated transcytosis for insulin or
transferrin, and active efflux
transporters such as p-glycoprotein. Active transport moieties may also be
conjugated to the
therapeutic or imaging compounds for use in the invention to facilitate
transport across the

51


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
epithelial wall of the blood vessel. Alternatively, drug delivery behind the
BBB is by intrathecal
delivery of therapeutics or imaging agents directly to the cranium, as through
an Ommaya
reservoir.
[0194] Where the therapeutic agents are locally administered in the brain, one
method for
administration of the therapeutic compositions of the invention is by
deposition into or near the
site by any suitable technique, such as by direct injection (aided by
stereotaxic positioning of an
injection syringe, if necessary) or by placing the tip of an Ommaya reservoir
into a cavity, or
cyst, for administration. Alternatively, a convection-enhanced delivery
catheter may be
implanted directly into the site, into a natural or surgically created cyst,
or into the normal brain
mass. Such convection-enhanced pharmaceutical composition delivery devices
greatly improve
the diffusion of the composition throughout the brain mass. The implanted
catheters of these
delivery devices utilize high-flow microinfusion (with flow rates in the range
of about 0.5 to 15.0
l/minute), rather than diffusive flow, to deliver the therapeutic composition
to the brain and/or
tumor mass. Such devices are described in U.S. Patent No. 5,720,720,
incorporated fully herein
by reference.
[0195] In some embodiments of the methods of treatment and methods of
administration
described herein, the methods include administering an effective amount of an
agent to promote
synapse formation. In some embodiments of the methods of treatment and methods
of
administration described herein, the methods include administering an
effective amount of an
agent to inhibit synapse formation. In some embodiments of the methods of
treatment and
methods of administration described herein, the methods include administering
an effective
amount of an agent to promote axonal and/or growth. In some embodiments of the
methods of
treatment and methods of administration described herein, the methods include
administering an
effective amount of an agent to inhibit axonal and/or dendritic growth. In
some embodiments, the
agent is an antagonist. In some embodiments, the agent is an agonist.
[0196] In some embodiments of the methods of treatment and methods of
administration
described herein, the methods comprise administering one or more modulators
described herein
(e.g., two modulators (e.g., an antagonist and an agonist)). In some
embodiments, the methods
include (a) administering an effective amount of a first agent to promote
axonal and/or dendritic
growth and (b) administering an effective amount of a second agent to promote
synapse
formation. In some embodiments, the first agent is an antagonist of a
thrombospondin and/or a
52


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
a28 subunit of a calcium channel. In some embodiments, the second agent is an
agonist of a
thrombospondin. In some embodiments, the first agent and second agent are
administered
sequentially. In some embodiments, the first agent and second agent are
administered separately.
In some embodiments, the first agent is administered less than about any of 1,
2, 3, 4, 5, 6, 7, 8,
9, or 10 days prior to the second agent. In some embodiments, the second agent
is administered
less than about any of 1, 3, 6, 9, 12, 18, 24, hours after first agent.
[0197] The methods of treatment and methods of administration described herein
including
agonists and/or antagonists of the present invention, are administered at a
dosage that modulates
synaptogenesis and/or axon growth while minimizing any side-effects. It is
contemplated that
compositions will be obtained and used under the guidance of a physician for
in vivo use. The
dosage of the therapeutic formulation will vary widely, depending upon the
nature of the disease,
the frequency of administration, the manner of administration, the clearance
of the agent from
the host, and the like.
[0198] The pharmaceutical compositions can be administered for prophylactic
and/or
therapeutic treatments. Toxicity and therapeutic efficacy of the active
ingredient can be
determined according to standard pharmaceutical procedures in cell cultures
and/or experimental
animals, including, for example, determining the LD50 (the dose lethal to 50%
of the population)
and the ED50 (the dose therapeutically effective in 50% of the population).
The dose ratio
between toxic and therapeutic effects is the therapeutic index and it can be
expressed as the ratio
LD50/ED50. Compounds that exhibit large therapeutic indices are preferred.
[0199] The data obtained from cell culture and/or animal studies can be used
in formulating
a range of dosages for humans. The dosage of the active ingredient typically
lines within a range
of circulating concentrations that include the ED50 with low toxicity. The
dosage can vary within
this range depending upon the dosage form employed and the route of
administration utilized.
[0200] The effective amount of a therapeutic composition described herein to
be given to a
particular patient will depend on a variety of factors, several of which will
be different from
patient to patient. Dosage of the agent will depend on the treatment, route of
administration, the
nature of the therapeutics, sensitivity of the patient to the therapeutics,
etc. Utilizing LD50 animal
data, and other information, a clinician can determine the maximum safe dose
for an individual,
depending on the route of administration. Utilizing ordinary skill, the
competent clinician will be
able to optimize the dosage of a particular therapeutic composition in the
course of routine

53


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
clinical trials. The compositions can be administered to the subject in a
series of more than one
administration. For therapeutic compositions, regular periodic administration
will sometimes be
required, or may be desirable. Therapeutic regimens will vary with the agent,
e.g. some agents
may be taken for extended periods of time on a daily or semi-daily basis,
while more selective
agents may be administered for more defined time courses, e.g. one, two three
or more days, one
or more weeks, one or more months, etc., taken daily, semi-daily, semi-weekly,
weekly, etc.
Pharmaceutically Acceptable Compositions and Formulations
[0201] Therapeutic agents, e.g. agonists or antagonists can be incorporated
into a variety of
formulations for therapeutic administration by combination with appropriate
pharmaceutically
acceptable carriers or diluents, and may be formulated into preparations in
solid, semi-solid,
liquid or gaseous forms, such as tablets, capsules, powders, granules,
ointments, solutions,
suppositories, injections, inhalants, gels, microspheres, and aerosols.
Pharmaceutical
compositions can include, depending on the formulation desired,
pharmaceutically-acceptable,
non-toxic carriers of diluents, which are defined as vehicles commonly used to
formulate
pharmaceutical compositions for animal or human administration. The diluent is
selected so as
not to affect the biological activity of the combination. Examples of such
diluents are distilled
water, buffered water, physiological saline, PBS, Ringer's solution, dextrose
solution, and
Hank's solution. In addition, the pharmaceutical composition or formulation
can include other
carriers, adjuvants, or non-toxic, nontherapeutic, nonimmunogenic stabilizers,
excipients and the
like. The compositions can also include additional substances to approximate
physiological
conditions, such as pH adjusting and buffering agents, toxicity adjusting
agents, wetting agents
and detergents.
[0202] The composition can also include any of a variety of stabilizing
agents, such as an
antioxidant for example. When the pharmaceutical composition includes a
polypeptide, the
polypeptide can be complexed with various well-known compounds that enhance
the in vivo
stability of the polypeptide, or otherwise enhance its pharmacological
properties (e.g., increase
the half-life of the polypeptide, reduce its toxicity, enhance solubility or
uptake). Examples of
such modifications or complexing agents include sulfate, gluconate, citrate
and phosphate. The
polypeptides of a composition can also be complexed with molecules that
enhance their in vivo

54


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
attributes. Such molecules include, for example, carbohydrates, polyamines,
amino acids, other
peptides, ions (e.g., sodium, potassium, calcium, magnesium, manganese), and
lipids.
[0203] Further guidance regarding formulations that are suitable for various
types of
administration can be found in Remington's Pharmaceutical Sciences, Mace
Publishing
Company, Philadelphia, PA, 17th ed. (1985). For a brief review of methods for
drug delivery,
see, Langer, Science 249:1527-1533 (1990).
[0204] For oral administration, the active ingredient can be administered in
solid dosage
forms, such as capsules, tablets, and powders, or in liquid dosage forms, such
as elixirs, syrups,
and suspensions. The active component(s) can be encapsulated in gelatin
capsules together with
inactive ingredients and powdered carriers, such as glucose, lactose, sucrose,
mannitol, starch,
cellulose or cellulose derivatives, magnesium stearate, stearic acid, sodium
saccharin, talcum,
magnesium carbonate. Examples of additional inactive ingredients that may be
added to provide
desirable color, taste, stability, buffering capacity, dispersion or other
known desirable features
are red iron oxide, silica gel, sodium lauryl sulfate, titanium dioxide, and
edible white ink.
Similar diluents can be used to make compressed tablets. Both tablets and
capsules can be
manufactured as sustained release products to provide for continuous release
of medication over
a period of hours. Compressed tablets can be sugar coated or film coated to
mask any unpleasant
taste and protect the tablet from the atmosphere, or enteric-coated for
selective disintegration in
the gastrointestinal tract. Liquid dosage forms for oral administration can
contain coloring and
flavoring to increase patient acceptance.
[0205] Formulations suitable for parenteral administration include aqueous and
non-
aqueous, isotonic sterile injection solutions, which can contain antioxidants,
buffers,
bacteriostats, and solutes that render the formulation isotonic with the blood
of the intended
recipient, and aqueous and non-aqueous sterile suspensions that can include
suspending agents,
solubilizers, thickening agents, stabilizers, and preservatives.
[0206] The components used to formulate the pharmaceutical compositions are
preferably
of high purity and are substantially free of potentially harmful contaminants
(e.g., at least
National Food (NF) grade, generally at least analytical grade, and more
typically at least
pharmaceutical grade). Moreover, compositions intended for in vivo use are
usually sterile. To
the extent that a given compound must be synthesized prior to use, the
resulting product is
typically substantially free of any potentially toxic agents, particularly any
endotoxins, which


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
may be present during the synthesis or purification process. Compositions for
parental
administration are also sterile, substantially isotonic and made under GMP
conditions.
[0207] Formulations may be optimized for retention and stabilization in the
brain. When the
agent is administered into the cranial compartment, it is desirable for the
agent to be retained in
the compartment, and not to diffuse or otherwise cross the blood brain
barrier. Stabilization
techniques include cross-linking, multimerizing, or linking to groups such as
polyethylene
glycol, polyacrylamide, neutral protein carriers, etc. in order to achieve an
increase in molecular
weight.
[0208] Other strategies for increasing retention include the entrapment of the
agent in a
biodegradable or bioerodible implant. The rate of release of the
therapeutically active agent is
controlled by the rate of transport through the polymeric matrix, and the
biodegradation of the
implant. The transport of drug through the polymer barrier will also be
affected by compound
solubility, polymer hydrophilicity, extent of polymer cross-linking, expansion
of the polymer
upon water absorption so as to make the polymer barrier more permeable to the
drug, geometry
of the implant, and the like. The implants are of dimensions commensurate with
the.size and
shape of the region selected as the site of implantation. Implants may be
particles, sheets,
patches, plaques, fibers, microcapsules and the like and may be of any size or
shape compatible
with the selected site of insertion.
[0209] The implants may be monolithic, i.e. having the active agent
homogenously
distributed through the polymeric matrix, or encapsulated, where a reservoir
of active agent is
encapsulated by the polymeric matrix. The selection of the polymeric
composition to be
employed will vary with the site of administration, the desired period of
treatment, patient
tolerance, the nature of the disease to be treated and the like.
Characteristics of the polymers will
include biodegradability at the site of implantation, compatibility with the
agent of interest, ease
of encapsulation, a half-life in the physiological environment.
[0210] Biodegradable polymeric compositions which may be employed may be
organic
esters or ethers, which when degraded result in physiologically acceptable
degradation products,
including the monomers. Anhydrides, amides, orthoesters or the like, by
themselves or in
combination with other monomers, may find use. The polymers will be
condensation polymers.
The polymers may be cross-linked or non-cross-linked. Of particular interest
are polymers of
hydroxyaliphatic carboxylic acids, either homo- or copolymers, and
polysaccharides. Included

56


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
among the polyesters of interest are polymers of D-lactic acid, L-lactic acid,
racemic lactic acid,
glycolic acid, polycaprolactone, and combinations thereof. By employing the L-
lactate or D-
lactate, a slowly biodegrading polymer is achieved, while degradation is
substantially enhanced
with the racemate. Copolymers of glycolic and lactic acid are of particular
interest, where the
rate of biodegradation is controlled by the ratio of glycolic to lactic acid.
The most rapidly
degraded copolymer has roughly equal amounts of glycolic and lactic acid,
where either
homopolymer is more resistant to degradation. The ratio of glycolic acid to
lactic acid will also
affect the brittleness of in the implant, where a more flexible implant is
desirable for larger
geometries. Among the polysaccharides of interest are calcium alginate, and
functionalized
celluloses, particularly carboxymethylcellulose esters characterized by being
water insoluble, a
molecular weight of about 5 kD to 500 kD, etc. Biodegradable hydrogels may
also be employed
in the implants of the subject invention. Hydrogels are typically a copolymer
material,
characterized by the ability to imbibe a liquid. Exemplary biodegradable
hydrogels which may
be employed are described in Heller in: Hydrogels in Medicine and Pharmacy, N.
A. Peppes ed.,
Vol. III, CRC Press, Boca Raton, Fla., 1987, pp 137-149.

Articles of Manufacture and Kits
[0211] The invention provides articles of manufacture comprising the
compositions,
formulations, and unit dosages described herein in suitable packaging for use
in the methods of
treatment and methods of administration described herein. Suitable packaging
for compositions
described herein are known in the art, and include, for example, vials (such
as sealed vials),
vessels (such as sealed vessels), ampules, bottles, jars, flexible packaging
(e.g., sealed Mylar or
plastic bags), and the like. These articles of manufacture may further be
sterilized and/or sealed.
[0212] The invention also provides a pharmaceutical pack or kit comprising one
or more
containers filled with one or more of the ingredients of the pharmaceutical
compositions of the
invention. Associated with such container(s) can be a notice in the form
prescribed by a
governmental agency regulating the manufacture, use or sale of pharmaceuticals
or biological
products, which notice reflects approval by the agency of manufacture, use or
sale for human
administration.
[0213] Instructions supplied in the kits of the invention are typically
written instructions on
a label or package insert (e.g., a paper sheet included in the kit), but
machine-readable

57


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
instructions (e.g., instructions carried on a magnetic or optical storage
disk) are also acceptable.
The instructions relating to the use of the nanoparticle compositions
generally include
information as to dosage, dosing schedule, and route of administration for the
intended treatment.
The kit may further comprise a description of selecting an individual suitable
or treatment.
[0214] The present invention further provides kits comprising compositions (or
unit dosages
forms and/or articles of manufacture) described herein and may further
comprise instruction(s)
on methods of using the composition, such as uses further described herein. In
some
embodiments, the kit of the invention comprises the packaging described above.
In other
embodiments, the kit of the invention comprises the packaging described above
and a second
packaging comprising a buffer. It may further include other materials
desirable from a
commercial and user standpoint, including other buffers, diluents, filters,
needles, syringes, and
package inserts with instructions for performing any methods described herein.

Methods for Screening
[0215] In some aspects of the invention, the invention provides methods for
screening a
candidate agent for activity in enhancing synaptogenesis, the method
comprising: a) measuring
binding of a candidate agent to an a28 polypeptide (e.g., a261 polypeptide) or
a thrombospondin
EGF-like domain; b) quantitating formation of synapses in a neural cell
culture in the presence of
the candidate agent if the candidate agent binds to the a28 polypeptide (e.g.,
a261 polypeptide)
or the thrombospondin EGF-like domain in step a), wherein an increased
formation of synapses
in the presence the candidate agent as compared to the formation of synapses
in the absence of
the candidate agent indicates that the candidate agent has the activity in
enhancing
synaptogenesis. The invention also provides methods for screening a candidate
agent for activity
in inhibiting synaptogenesis, the method comprising: a) measuring binding of a
candidate agent
to an a25 polypeptide (e.g., a261 polypeptide) or a thrombospondin EGF-like
domain; b)
quantitating formation of synapses in a neural cell culture in the presence of
the candidate agent
and a thrombospondin agonist if the candidate agent binds to the a28
polypeptide (e.g., (X281
polypeptide) or the thrombospondin EGF-like domain in step a), wherein a
decreased formation
of synapses in the presence the candidate agent as compared formation of
synapses in the
absence of the candidate agent indicates that the candidate agent has the
activity in inhibiting
synaptogenesis.

58


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
[0216] Candidate agents are screened for the ability to modulate
synaptogenesis, which
agents may include candidate thrombospondin derivatives, variants, fragments,
mimetics,
agonists and antagonists, and/or GABA analogs and mimetics. Agents of interest
may be
screened against calcium channel subunit a26 (e.g., subunit a26 1), and/or a
polypeptide
comprising at least one thrombospondin EGF-like domain. Such compound
screening may be
performed using an in vitro model, a cell expressing the polypeptide,
including a genetically
altered cell or animal, or purified protein. A wide variety of assays may be
used for this purpose.
In one embodiment, compounds that are predicted to be antagonists or agonists
of
synaptogenesis are initially tested for binding to calcium channel subunit a26
(e.g., calcium
channel subunit a251), or for interacting with a thrombospondin EGF-like
domain. The
compounds may then be further tested for functional activity in a biological
model, e.g. an in
vitro culture system, as described below, an animal model, etc.
[0217] For example, candidate agents may be identified by known pharmacology,
by
structure analysis, by rational drug design using computer based modeling, by
binding assays,
and the like. Various in vitro models may be used to determine whether a
compound binds to, or
otherwise affects thrombospondin activity. Such candidate compounds are used
to contact
neurons in an environment permissive for synaptogenesis. Such compounds may be
further
tested in an in vivo model for enhanced synaptogenesis.
[0218] Synaptogenesis is quantitated by administering the candidate agent to
neurons in
culture, and determining the presence of synapses in the absence or presence
of the agent. In one
embodiment of the invention, the neurons are a primary culture, e.g. of RGCs.
Purified
populations of RGCs are obtained by conventional methods, such as sequential
immunopanning.
The cells are cultured in suitable medium, which will usually comprise
appropriate growth
factors, e.g. CNTF; BDNF; etc. As a positive control, soluble thrombospondin,
e.g. TSP1, TSP2,
etc. may be added to certain wells. The neural cells, e.g. RCGs, are cultured
for a period of time
sufficient allow robust process outgrowth and then cultured with a candidate
agent for a period
of about 1 day to 1 week, to allow synapse formation. For synapse
quantification, cultures are
fixed, blocked and washed, then stained with antibodies specific synaptic
proteins, e.g.
synaptotagmin, etc. and visualized with an appropriate reagent, as known in
the art. Analysis of
the staining may be performed microscopically. In one embodiment, digital
images of the
fluorescence emission are with a camera and image capture software, adjusted
to remove unused

59


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
portions of the pixel value range and the used pixel values adjusted to
utilize the entire pixel
value range. Corresponding channel images may be merged to create a color
(RGB) image
containing the two single-channel images as individual color channels. Co-
localized puncta can
be identified using a rolling ball background subtraction algorithm to remove
low-frequency
background from each image channel. Number, mean area, mean minimum and
maximum pixel
intensities, and mean pixel intensities for all synaptotagmin, PSD-95, and
colocalized puncta in
the image are recorded and saved to disk for analysis.
[0219] In some embodiments, candidate agents encompass numerous chemical
classes,
though typically they are organic molecules, preferably small organic
compounds having a
molecular weight of more than 50 and less than about 2,500 daltons. Candidate
agents comprise
functional groups necessary for structural interaction with proteins,
particularly hydrogen
bonding, and typically include at least an amine, carbonyl, hydroxyl or
carboxyl group,
preferably at least two of the functional chemical groups. The candidate
agents often comprise
cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic
structures substituted
with one or more of the above functional groups. Candidate agents are also
found among
biomolecules including peptides, saccharides, fatty acids, steroids, purines,
pyrimidines,
derivatives, structural analogs or combinations thereof. Generally a plurality
of assay mixtures
are run in parallel with different agent concentrations to obtain a
differential response to the
various concentrations. Typically one of these concentrations serves as a
negative control, i.e. at
zero concentration or below the level of detection.
[0220] Candidate agents are obtained from a wide variety of sources including
libraries of
synthetic or natural compounds. For example, numerous means are available for
random and
directed synthesis of a wide variety of organic compounds and biomolecules,
including
expression of randomized oligonucleotides and oligopeptides. Alternatively,
libraries of natural
compounds in the form of bacterial, fungal, plant and animal extracts are
available or readily
produced. Additionally, natural or synthetically produced libraries and
compounds are readily
modified through conventional chemical, physical and biochemical means, and
may be used to
produce combinatorial libraries. Known pharmacological agents may be subjected
to directed or
random chemical modifications, such as acylation, alkylation, esterification,
amidification, etc. to
produce structural analogs. Test agents can be obtained from libraries, such
as natural product
libraries or combinatorial libraries, for example.



CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
[0221] Libraries of candidate compounds can also be prepared by rational
design. (See
generally, Cho et al., Pac. Symp. Biocompat. 305-16, 1998); Sun et al., J.
Comput. Aided Mol.
Des. 12:597-604, 1998); each incorporated herein by reference in their
entirety). For example,
libraries of phosphatase inhibitors can be prepared by syntheses of
combinatorial chemical
libraries (see generally DeWitt et al., Proc. Nat. Acad. Sci. USA 90:6909-13,
1993; International
Patent Publication WO 94/08051; Baum, Chem. & Eng. News 72:20-25, 1994;
Burbaum et al.,
Proc. Nat. Acad. Sci. USA 92:6027-31, 1995; Baldwin et al., J. Am. Chem. Soc.
117:5588-89,
1995; Nestler et al., J. Org. Chem. 59:4723-24, 1994; Borehardt et al., J. Am.
Chem. Soc.
116:373-74, 1994; Ohlmeyer et al., Proc. Nat. Acad. Sci. USA 90:10922-26, all
of which are
incorporated by reference herein in their entirety.)
[0222] A "combinatorial library" is a collection of compounds in which the
compounds
comprising the collection are composed of one or more types of subunits.
Methods of making
combinatorial libraries are known in the art, and include the following: U.S.
Patent Nos.
5,958,792; 5,807,683; 6,004,617; 6,077,954; which are incorporated by
reference herein. The
subunits can be selected from natural or unnatural moieties. The compounds of
the combinatorial
library differ in one or more ways with respect to the number, order, type or
types of
modifications made to one or more of the subunits comprising the compounds.
Alternatively, a
combinatorial library may refer to a collection of "core molecules" which vary
as to the number,
type or position of R groups they contain and/or the identity of molecules
composing the core
molecule. The collection of compounds is generated in a systematic way. Any
method of
systematically generating a collection of compounds differing from each other
in one or more of
the ways set forth above is a combinatorial library.
[0223] A combinatorial library can be synthesized on a solid support from one
or more solid
phase-bound resin starting materials. The library can contain five (5) or
more, preferably ten (10)
or more, organic molecules that are different from each other. Each of the
different molecules is
present in a detectable amount. The actual amounts of each different molecule
needed so that its
presence can be determined can vary due to the actual procedures used and can
change as the
technologies for isolation, detection and analysis advance. When the molecules
are present in
substantially equal molar amounts, an amount of 100 picomoles or more can be
detected.
Preferred libraries comprise substantially equal molar amounts of each desired
reaction product

61


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
and do not include relatively large or small amounts of any given molecules so
that the presence
of such molecules dominates or is completely suppressed in any assay.
[0224] Combinatorial libraries are generally prepared by derivatizing a
starting compound
onto a solid-phase support (such as a bead). In general, the solid support has
a commercially
available resin attached, such as a Rink or Merrifield Resin. After attachment
of the starting
compound, substituents are attached to the starting compound. Substituents are
added to the
starting compound, and can be varied by providing a mixture of reactants
comprising the
substituents. Examples of suitable substituents include, but are not limited
to, hydrocarbon
substituents, e.g. aliphatic, alicyclic substituents, aromatic, aliphatic and
alicyclic-substituted
aromatic nuclei, and the like, as well as cyclic substituents; substituted
hydrocarbon substituents,
that is, those substituents containing nonhydrocarbon radicals which do not
alter the
predominantly hydrocarbon substituent (e.g., halo (especially chloro and
fluoro), alkoxy,
mercapto, alkylmercapto, nitro, nitroso, sulfoxy, and the like); and hetero
substituents, that is,
substituents which, while having predominantly hydrocarbyl character, contain
other than carbon
atoms. Suitable heteroatoms include, for example, sulfur, oxygen, nitrogen,
and such substituents
as pyridyl, furanyl, thiophenyl, imidazolyl, and the like. Heteroatoms, and
typically no more than
one, can be present for each carbon atom in the hydrocarbon-based
substituents. Alternatively,
there can be no such radicals or heteroatoms in the hydrocarbon-based
substituent and, therefore,
the substituent can be purely hydrocarbon.
[0225] Compounds that are initially identified by any screening methods can be
further
tested to validate the apparent activity. The basic format of such methods
involves administering
a lead compound identified during an initial screen to an animal that serves
as a model for
humans and then determining the effects on synaptogenesis. The animal models
utilized in
validation studies generally are mammals. Specific examples of suitable
animals include, but are
not limited to, primates, mice, and rats.

EXAMPLES
[0226] The following examples are put forth so as to provide those of ordinary
skill in the
art with a complete disclosure and description of how to make and use the
subject invention, and
are not intended to limit the scope of what is regarded as the invention.
Efforts have been made
to ensure accuracy with respect to the numbers used (e.g. amounts,
temperature, concentrations,

62


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
etc.) but some experimental errors and deviations should be allowed for.
Unless otherwise
indicated, parts are parts by weight, molecular weight is average molecular
weight, temperature
is in degrees centigrade; and pressure is at or near atmospheric.
Example 1
[0227] The number of synapses between CNS neurons in culture is profoundly
enhanced by
a soluble signal secreted by astrocytes, which are identified herein as
thrombospondins (TSPs),
which are a necessary and sufficient component of the synapse-promoting
activity of astrocyte-
conditioned medium. TSPs induce ultrastructurally normal synapses that are
presynaptically
active but postsynaptically inactive. In vivo, TSPs are concentrated in
astrocytes and at synapses
throughout the developing brain, and mice deficient in both TSP 1 and its
ortholog TSP2 have a
significant decrease in synapse number.
[0228] TSPs are large oligomeric extracellular matrix proteins, about 500 kD,
that mediate
cell-cell and cell-matrix interactions by binding an array of membrane
receptors, other
extracellular matrix proteins, and cytokines. There are five TSPs, each
encoded by a separate
gene. Although several TSPs are expressed in the brain, the functions of these
TSPs are
unknown. TSPI and TSP2 are closely related trimeric proteins that share the
same set of
structural and functional domains. TSP4, which is pentameric and has a
different domain
structure from TSP1 and TSP2, is present in the adult nervous system where it
is localized to
some CNS synapses as well as the neuromuscular junction.
[0229] All 5 TSP isoforms have strong synapse inducing activity as a result of
sharing a
common EGF like domain. Using this domain, we have identified that TSP induces
synapse
formation through a novel interaction with a widely expressed transmembrane
neuronal cell
surface molecule, calcium channel subunit alpha2 deltal, which has not been
previously linked
to synapse formation.
[0230] Gabapentin, which is used as antiepileptic and anti-neuropathic pain
agent, has been
previously shown to bind to this receptor, but its mechanism of action has
long been a mystery.
We have shown that Gabapentin in culture is able to specifically inhibit the
synapse forming
activity of thrombospondin or its domain. This action is specific for
thrombospondin as
Gabapentin does not inhibit the synapse formation induced by another astrocyte
secreted
synaptogenic protein Hevin.

63


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
[0231] As shown in Figure lA, all TSPs are extracellular multimeric,
multidomain,
calcium-binding glycoproteins that function at cell surfaces and in the
extracellular matrix
milieu. The thrombospondin gene family is divided into two subgroups. Subgroup
A
thrombospondins includes TSPI and 2, which are trimeric and have larger N
terminal domains
(black oval) and which have three additional properdin like repeats
(rectangles). Subgroup B
TSPs are pentameric and lack the properdin like repeats.
[0232] Previously, it was shown that Subgroup A TSP1 and 2 are synaptogenic
(Christopherson et. al. Cell, 2005). Here it is shown that pentameric Subgroup
B
thrombospondins are also synaptogenic. Immunostaining of RGCs for co-
localization of
presynaptic Synaptotagmin and postsynaptic PSD-95 showed few colocalized
synaptic puncta in
the absence of astrocytes (B) but many were found in the presence of a feeding
layer of
astrocytes (Figure 1 C).
[0233] RGCs cultures with conditioned media from COS7 cells transfected with
TSP3
overexpression vector (Figure 1 D) or with purified TSP4 (Figure lE) or TSP5
(Figure 1 F)
formed many synapses, which are observed as co-localized pre and post-synaptic
puncta.
Quantification of the effects of astrocytes and TSPs on synaptic puncta is
shown in Figure 1 G.
Astrocytes and TSP1, TSP4 or TSP5 both significantly increased the number of
co-localized
synaptic puncta/cell over RGCs alone (Control). Shown in Figure 1H, culturing
RGCs with the
conditioned media from COS7 cells transfected with TSP3 overexpression vector
significantly
increased the number of co-localized synaptic puncta/cell when compared with
RGCs cultured
with conditioned media from COS7 cells transfected with empty vector (Control)
(*p<0.05,
n=20, error bars indicate SEM values).
[0234] Shown in Figure 2A is the thrombospondin domain structure. TSP1 and 2
consist of
a heparin-binding N terminal domain (C), a linker with homology to procollagen
(PC), three
TSP-type-1 (properdin) repeats, three EGF-like (TSP- type-2) repeats, seven
TSP type-3
(calcium binding) repeats, and a cell-binding carboxyl-terminal domain (C).
[0235] The quantification of the effects of TSP 1(B) and TSP2 (C) domains on
the number
of synapses formed by RGCs in vitro is shown in Figure 2B. Astrocytes and
purified TSPI
(trimeric-8nM) increased the number of synapses formed by RGCs significantly
when compared
to RGCs cultured alone. Interestingly TSPI trunctation constructs (monomeric-
20nM) that
contained the EGF-like repeats (blue) could also significantly increase the
synapse number.

64


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
[0236] Shown in Figure 2C, similarly purified recombinant TSP2 domains
(monomeric-
20nM) that contained the EGF-like repeats of TSP2 increased the number of
synapses formed by
RGCs significantly when compared to RGCs cultured alone. Interestingly a
construct that
contained only the third EGF-like domain together with the C-terminal end of
TSP2 was also
synaptogenic. However, the third EGF-like repeat (blue) alone did not
significantly increase the
synapse number. (Figures 2D, E). Antibodies against EGF like repeats of TSPs
can block their
synaptogenic effect. RGCs cultured with astrocytes or with the recombinant
TSP1 (Figure 2D) or
TSP2 (Figure 2E) truncation constructs that contained the third properdin
repeats with the three
EGF-like domains (will be referred as EGF-like domain here on) formed many
more synapses
when compared to RGCs cultured alone.
[0237] Interestingly, a monoclonal antibody A4.1 (Neomarkers) that binds to
the third EGF-
like domain of TSPs blocked the synaptogenic effect of both TSP 1 and TSP2
synaptogenic
constructs. Another monoclonal antibody C6.7 (Neomarkers) that binds to the
second EGF-like
repeats of TSPs did not affect the synaptogenic function of TSP 1 construct
however if was able
to block the synaptogenic TSP2 domain. These data show that the synaptogenic
effect of TSPs is
mediated through the EGF-like repeats of TSPs and most likely through an
interaction mapped to
the third EGF-like domain. (*p<0.05, n=20, error bars indicate SEM values).
[0238] Figure 3A shows immunostaining of RGCs for co-localization of
presynaptic
Synaptotagmin and postsynaptic PSD-95 showed few co-localized synaptic puncta
when
cultured alone but many in the presence of purified TSP1 or the recombinant
TSP2 EGF-like
domains.
[0239] Gabapentin (GBP, 32 M) a drug that binds to calcium channel subunit
alpha2delta
blocked the synaptogenic effect of TSPI or the TSP2 EGF-like domains, as shown
in Figure 3B.
Figure 3C, quantification of the effects of Gabapentin on TSP induced synapse
formation.
Gabapentin (32 M) addition to TSP1 or TSP2 EGF-like domains reduces the number
of
synapses formed by RGCs down to background levels. Figure 3D, gabapentin
blocks the
synaptogenic effect of astrocyte conditioned media (ACM). RGCs cultured in the
presence of rat
or mouse ACM formed 5-10 fold higher number of synapses when compared to RGCs
cultured
alone. Addition of Gabapentin (32 M) diminished the synaptogenic effect of
both rat and mouse
ACM.



CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
[0240] Figure 3E, gabapentin blocks formation of new synapses but can not
break down
already-formed synapses or synapses formed by another synaptogenic protein
Hevin. 3 DIV
RGCs were treated with purified recombinant TSP2 EGF-like domains (20nM) or
Hevin (30nM)
for 9 days in vitro. Both the EGF-like domains and Hevin increased the number
of synapses
formed significantly when compared to RGCs cultured alone. Similar to (C)
addition of
Gabapentin (GBP) together with EGF-like domains diminished its synaptogenic
effect. However,
addition of Gabapentin to RGCs treated with EGF-like domains at day 6 for an
additional 3 days
did not decreased the synapse number indicating that Gabapentin did not
breakdown already
formed synapses, but rather blocked formation of new synapses. Gabapentin
addition to Hevin,
another astrocyte secreted protein that can induce formation of structural
synapses, did not affect
Hevin's synaptogenic function regardless of the time it was added. (*p<0.05,
n=20, error bars
indicate SEM values).
Methods
[0241] Purification and culture of RGCs. RGCs were purified by sequential
immunopanning to greater than 99.5% purity from P5 Sprague-Dawley rats
(Simonsen Labs,
Gilroy, CA), as previously described (Barres et. al. (1988) Neuron 9, 791).
Approximately
30,000 RGCs were cultured per well in 24-well plates (Falcon) on glass
(Assistant) or Aclar 22C
(Allied Signal) coverslips coated with poly-D-lysine (10 g/ml) followed by
laminin (2 g/ml).
RGCs were cultured in 600 l of serum-free medium, modified from Bottenstein
and Sato
(1979), containing Neurobasal (Gibco), bovine serum albumin, selenium,
putrescine, triiodo-
thyronine, transferrin, progesterone, pyruvate (1 mM), glutamine (2 mM), CNTF
(10 ng/ml),
BDNF (50 ng/ml), insulin (5 g/ml), and forskolin (10 M). Recombinant human
BDNF and
CNTF were generously provided by Regeneron Pharmaceuticals.
[0242] Purified human platelet TSP1 was from either Sigma or Haematologic
Technologies
with similar results. Recombinant TSP2 was purified from serum-free medium
conditioned by
baculovirus-infected insect cells expressing mouse TSP2. Since purified TSP1
is readily
available, we used this as the source of TSP in our experiments unless
otherwise stated TSPs
were used at a concentration of 5 g/ml unless otherwise specified. RCGs were
cultured for 4
days to allow robust process outgrowth and then cultured with TSPs for an
additional 6 days. All
other reagents were obtained from Sigma.

66


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
[0243] Preparation of astrocytes and ACM. Cortical glia were prepared as
described by
McCarthy, J. de Vellis, J. Cell Biol. 85, 890 (1980). Briefly, postnatal day1-
2 cortices were
papain-digested and plated in tissue culture flasks (Falcon) in a medium that
does not allow
neurons to survive (Dulbecco's Modified Eagle Medium, fetal bovine serum
(10%), penicillin
(100 U/ml), streptomycin (100 g/ml), glutamine (2 mM) and Na-pyruvate (1 mM).
After 4 days
non-adherent cells were shaken off of the monolayer and cells were incubated
another 2-4 days
to allow monolayer to refill. Medium was replaced with fresh medium containing
AraC (10 M)
and incubated for 48 hours. Astrocytes were trypsinized and plated onto 24-
well inserts (Falcon,
1.0 m) or 10 cm tissue culture dishes.
[0244] For preparation of ACM, confluent cultures of astrocytes in 10 cm
dishes were
washed 3X in PBS and fed with 10 mls RGC medium (without CNTF, BDNF or
forskolin).
ACM was harvested after 4-6 days of conditioning, filtered through a 0.2 m
syringe filter and
concentrated l Ox through a 5 KD molecular weight cut-off centrifuge
concentrator (Millipore),
unless otherwise indicated. ACM was used at a final concentration of 5X unless
otherwise
indicated. RCGs were cultured for 4 days to allow robust process outgrowth and
then cultured
with ACM or an astrocyte-feeding layer for an additional 6 days.
[0245] Synaptic assays. For synapse quantification, cultures were fixed for 7
min in 4%
paraformaldehyde (PFA), washed 3X in phosphate buffered saline (PBS) and
blocked in 100 L
of blocking buffer (50% Antibody Buffer (0.5% bovine serum albumin, 0.5 %
Triton X-100, 30
mM NaPO4, 750 mM NaCl, 5% normal goat serum, and 0.4% NaN3, pH 7.4), 50% goat
serum
(NGS), 0.1% Triton-X) for 30 min. After blocking, cover slips were washed 3X
in PBS and
100 L of primary antibody solution was added to each cover slip, consisting of
rabbit anti-
synaptotagmin (cytosolic domain, Synaptic Systems) and mouse anti-PSD-95 (6G6-
IC9 clone,
Affinity Bio Reagents) diluted 1:500 in antibody buffer. Coverslips were
incubated overnight at
4 C, washed 3X in PBS, and incubated with 100 L of secondary antibody
solution containing
Alexa-594 conjugated goat anti-rabbit and Alexa-488 conjugated goat anti-mouse
(Molecular
Probes) diluted 1:1000 in antibody buffer. Following incubation for 2 h at
room temperature,
coverslips were washed five times in PBS and mounted in Vectashield mounting
medium with
DAPI (Vector Laboratories Inc) on glass slides (VWR Scientific). For
presynaptic activity assay,
rabbit synaptotagmin antiserum was generated by immunization with a peptide
corresponding to
the N-terminal luminal portion of synaptotagmin. This serum was added at 1:500
to live cultures

67


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
and incubated for 6 hours. Cells were then washed 3X in DPSB, fixed and
stained as above,
except for the omission of synaptotagmin antibody from the primary antibody
solution.
[0246] Mounted coverslips were imaged using Nikon Diaphot and Eclipse
epifluorescence
microscopes (Nikon). Healthy cells that were at least 2 cell diameters from
their nearest neighbor
were identified and selected at random by eye using DAPI fluorescence. 8-bit
digital images of
the fluorescence emission at both 594 nm and 488 nm were recorded for each
selected cell using
a cooled monochrome CCD camera and SPOT image capture software (Diagnostic
Instruments,
Inc). Each single-channel image was adjusted to remove unused portions of the
pixel value range
and the used pixel values were adjusted appropriately to utilize the entire
pixel value range.
Corresponding channel images were then merged to create a color (RGB) image
containing the
two single-channel images as individual color channels. These manipulations
were performed
automatically using the custom software package SpotRemover ( 2001 Barry
Wark).
[0247] Colocalized puncta were identified using a custom-written plug-in. Full
documentation of the puncta-counting algorithm is available in the "Puncta
Analyzer" plug-in's
source code. Briefly, the rolling ball background subtraction algorithm was
used to remove low-
frequency background from each image channel. The puncta were "masked" in the
single-
channel image by thresholding the image so that only legitimate synaptic
puncta remained above
threshold. ImageJ's "Particle Analyzer" plug-in was then used to identify and
characterize
puncta within each channel. Puncta in different color channels were defined as
colocalized if the
centers of two circles, centered at the puncta's centroids and with areas
equal to the puncta's
area, were less than the larger of the two circle's radius apart. Number, mean
area, mean
minimum and maximum pixel intensities, and mean pixel intensities for all
synaptotagmin, PSD-
95, and colocalized puncta in the image were recorded and saved to disk for
later analysis.
[0248] Immunohistochemistry. Brain sections were dried 30 min at 37 C followed
by
application of blocking buffer. Slides were washed 3 X 5 min in PBS. Primary
used were diluted
into antibody buffer as follows: TSP 1(P 10, mouse monoclonal, Immunotech,
1:200 or Ab 8,
Neomarkers, rabbit, 1:200), synaptotagmin (rabbit polyclonal, Synaptic
Systems, 1:500), ezrin
(monoclonal 3C12, Neomarkers, 1:200), SV2 (hybridoma supernatant,
Developmental Studies
Hybridoma Bank, 1:30), Bassoon (Stressgen, 1:400), PSD-95 (monoclonal 6G6-1
C9, Affinity
Bioreagents, 1:250) and incubated overnight at 4 C followed by 3X washes in
PBS. Secondary

68


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
Alexa-conjugated antibodies (Molecular Probes) were added at 1:1000 for 2
hours at RT. Slides
were washed 3X in PBS and mounted in Vectashield plus DAPI.
[0249] Confocal analysis of synapse number. Images of immunostained brains
were
collected on a Leica SPS SP2 AOBS confocal microscope. Optical sections were
line-averaged
and collected at 0.28 M intervals. Gain, threshold, and black levels were
individually adjusted
per section to cover the same range of pixel values, or were set for the WT
sections and kept
constant for all sections. In both cases equivalent results were obtained for
the relative number of
synapses in WT or KO animals. Stacks of 20 optical sections were quantified
for synapse
number by projecting a series of 5 optical sections, a number empirically
determined to optically
section the entirety of most synaptic puncta, and counting the number of
synapses in each
projection volume. Synapses were automatically counted using the ImageJ puncta
analyzer
program and the accuracy of the counts confirmed by counting by hand. N= 6
hemispheres for
P8 WT and KO and N = 10 hemispheres and for P21 WT and KO. On average 3 stacks
per
hemisphere were obtained yielding a total of 18 stacks (72 optical sections)
for synaptic puncta
analysis for P8 brains and a total of 30 stacks (120 optical sections) for
synaptic puncta analysis
for P21 brains.
Example 2-The calcium channel subunit a281 is the neuronal TSP receptor
involved in
synapse formation
[0250] To determine whether a281 plays a role in TSP-induced synapse formation
in vitro,
we overexpressed a26l in RGCs and determined whether TSP-induced synapse
formation was
affected. RGCs which overexpress a281 formed twice as many synapses in
response to SD2 as
did RGCs transfected with GFP alone (Figure 4A) indicating that a281
overexpression enhances
TSP-induced synapse formation. a251 overexpression alone was not sufficient to
induce synapse
formation in the absence of TSP indicating that TSP-a281 interaction is
required for the
initiation of synapse formation.
[0251] To further determine whether a281 is required for TSP induced synapse
formation, a
small interfering RNA (siRNA) knockdown approach was used. A siRNA pool
specific for rat
a281 significantly reduced the expression of rat a251 in transfected HEK293
cells (Figure 4B).
RGCs were transfected with this siRNA pool against rat a281 or with siRNA
control pools.
Knockdown of a281 strongly inhibited astrocyte or TSP induced synapse
formation in vitro
(Figure 4C, 4D and data not shown) whereas neither the non-targeting control
siRNA pool

69


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
(siControl) nor the targeting siRNA pool against the rat integrin (31 protein,
which is another TSP
receptor present at RGC synapses, affected synapse formation (Figure 4C, 4D).
These results
demonstrate that a251 is necessary for TSP and astrocyte induced synapse
formation in vitro.
Because only synapses on GFP-expressing cells were counted in both the a251
overexpression
and knockdown experiments, these results also indicate that a281 is acting on
the postsynaptic
side and is necessary for the cells ability to receive synapses.
Example 3-a281 promoted synapse formation does not depend on calcium channel
function
or number
[0252] In order to find out whether a261 promotes synapse formation through
its ability to
modulate biophysical properties of calcium channels, whether the 81 subunit
could still enhance
TSP-induced synapse formation was tested. To do so, 61 was overexpressed in
RGCs and
analyzed for its effect on SD2-induced synapse formation. Overexpression of 81
subunit did not
mimic the effect of full-length a261. On the contrary, the 81 subunit acted as
a dominant
negative construct and led to inhibition of SD2-induced synapse formation
(Figure 4E).
Similarly, 81 subunit expression also blocked astrocyte-induced synapse
formation (data not
shown). These results indicate that a281 does not mediate its effect on
synapse formation
through modulating biophysical properties of the calcium channel. In addition,
the enhancement
of TSP-induced synapse formation by a281 requires the presence of the a2
subunit of the
receptor, which contains the VWF-A domain. Furthermore, the dominant negative
effect of the 8
subunit suggests that the S subunit is required for transmitting the synapse-
inducing signal upon
TSP-a2 interaction.
102531 Further investigation was conducted to determine whether the role of
a261 in
synapses formation is linked to its ability to increase calcium currents via
increasing surface
calcium channel number. Previously, we found that astrocytes increase the
total number of
calcium channels at the RGC cell surface (Ullian et al., Science 291:657-661,
2001). This
increase is small and concurrent with synapse formation rather than preceding
it, making it
unlikely to be the cause of synapse formation. To directly test whether
voltage gated calcium
channel (VGCC) function was required for synapse formation, the L-type calcium
channel
blockers nimodipine and nifedipine were added to the RGC culture medium to
determine if this
decreased SD2-induced synapse formation (Figure 5A). Blocking L-type channel
function,



CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
which accounts for the majority of the calcium currents in RGCs (Ullian et
al., Science 291:657-
661, 2001) and is predominantly postsynaptic, had no effect on TSP-induced
synapse formation,
even though RGC survival was negatively affected by the presence of these
drugs. Similarly
blockers for pre-synaptic N and P/Q type channels (Conotoxin GV 1A, Conotoxin
MVIIA,
Agatoxin IVA and Conotoxin MVIIC) did not block TSP-induced synapse formation
(data not
shown).
[02541 Finally, investigation was conducted to determine whether increasing
postsynaptic
L-type calcium channel expression in RGCs would enhance synapse formation in a
similar way
to overexpression of a281. To do so, L-type calcium channel a 1 C and 0
subunits were
overexpressed in RGCs and analyzed for their effect on astrocyte induced
synapse formation.
Overexpression of a 1 and (3 subunits had no positive or negative effect on
synapse formation in
the presence or absence of astrocytes (Figure 5B). These results suggest that
neither calcium
channel function, nor an increase in calcium channel number, is the driving
force behind TSP-
induced synapse formation, however, a261-VGCC interaction may still be
important for the
signaling events leading to the initiation of TSP-induced synapse formation.
Together these
observations providing further evidence that the a28 1-TSP interaction may
induces synapse
formation through a yet unknown function of a28 1.

Example 4-Overexpression of a281 in neurons enhances synapse formation in vivo
[02551 a251 knockouts are early embryonic lethal; therefore, it is difficult
to test the effect
of a281 on synapse formation in vivo. As an alternative approach, synapse
number was
examined in mice that overexpress a281 specifically in neurons, under the
control of the Thyl
promoter (Li et al., Pain 125:20-34, 2006). These transgenic mice are
hypersensitive to
mechanical and thermal stimulation due to spinal hyperexcitability and have
elevated levels of
a281 protein throughout the CNS. We investigated whether these mice have
higher levels of
excitatory synapses in the cortex. Sagital brain sections from 21-day-old (P2
1) transgenic and
wildtype littermate mice were co-immunostained with antibodies directed
against postsynaptic
density protein 95 (PSD95) and either the pre-synaptic vesicular glutamate
transporter 1 or 2
(VGlutl and VGlut2, respectively). The number of co-localized pre and
postsynaptic puncta was
quantified to determine the synaptic density in the cortices of these mice.
Transgenic mice
overexpressing a281 had significantly higher numbers of VGlut2 positive
excitatory synapses in
the cortex when compared with the littermate wildtype controls (Figure 6A and
B). On the other

71


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
hand, the number of VGlutl positive excitatory synapses was similar for both
the transgenic and
the wildtype mice (Figure 6C and D). The observation that a251 overexpression
specifically
increases VG1u5 positive synapses is interesting. One possible explanation is
that in these mice
the a261 transgene is specifically overexpressed in neurons that establish
VGlut2 positive
synapses. In conclusion, the increase in the number of VGlut2/PSD95 synapses
in a281
overexpressing mice shows that a261 plays a role in promoting excitatory
synapse formation in
the brain.
Example 5-Gabapentin, a high affinity ligand for a281, strongly inhibits TSP
and astrocyte
induced synapse formation
[0256] a261 is a high affinity receptor for gabapentin. In order to determine
whether
gabapentin (GBP) affects TSP or astrocyte induced synapse formation, RGCs were
cultured with
SD2 or ACM in the presence or absence of GBP (32 M). GBP strongly inhibited
both TSP and
astrocyte -induced synapse formation (Figure 7A-C). GBP was not toxic to the
RGCs (RGC
survival was 62.0 2.1 % in control culture media versus 66.5 4.1 % in
culture media
containing GBP). Similarly, GBP did not affect neurite outgrowth (data not
shown).
Investigation was conducted to determine whether GBP could dissolve synapses
that had already
formed. RGCs were cultured with SD2 for 5 days to allow synapses formation and
then added
GBP for an additional day. Although GBP completely inhibited synapse formation
induced by
SD2 when it was present for the entire 6-day culture period, when GMP added
SD2 for only the
last 24 hours synapse formation was not affected (Figure 7B). Thus GBP
powerfully blocks new
synapse formation induced by TSP and astrocytes, but does not dissolve already
formed
synapses. Interestingly GABA, an inhibitory neurotransmitter that binds to
a281 with much
lower affinity (IC50=650 M, (Suman-Chauhan et al., Eur. J. of Pharmacol.
244:293-301, 1993),
also blocked TSP induced synapse formation when used at high concentrations
(Figure 8).
[02571 To determine whether GBP similarly blocks synapse formation in vivo,
neonatal
mice were injected with either GBP or saline, for the first postnatal week,
which coincides with
the initiation of synapse formation in the brain (see Experimental
Procedures). Analysis was
carried out to determine whether GBP-injected mice had a reduced number of
glutamatergic
excitatory synapses in the cortex. At this age P7 glutamatergic synapses in
the cortex are
predominantly VGlut2 positive (Miyazaki et al., The Eur. J. of Neuroscience,
17:2563-2572,
2003). Therefore, co-immunostaining of sagital brain sections from (P7) saline
or GBP-injected,

72


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
mice was carried out with antibodies against VGlut2 and PSD95 and the number
of co-localized
pre- and post-synaptic puncta was quantified to determine synaptic density in
the cortex of these
mice. There was a significant decrease in the density of excitatory synapses
in the cerebral cortex
of the GBP injected mice in comparison to saline injected control mice (Figure
7D). This
difference was mainly due to a severe decrease in synapse number in half of
the GBP injected
animals. In this half of the mice that responded to GBP, the VGlut2/PSD95
synaptic densities
went down profoundly to less than 10% of the saline injected values, although
there was no
apparent effect on the number of neurons. GBP injection affected both VGlut2
and PSD95
puncta by reducing their number, size, and co-localization (Figure 7E) similar
to its effect on
synaptic puncta in vitro (Figure 7A). The other half of the GBP injected group
had a similar
synaptic density as the saline injected controls. It is interesting that the
effect of GBP in vivo is
an "all or none" effect rather than a gradual decrease in synapse number, and
that only 50% of
the mice responded to GBP injection. This could be due to a critical threshold
concentration of
GBP necessary to be effective in blocking synapse formation, which might only
be achieved in
some of the mice. In three repeated experiments, however, we observed only
half of the mice
were affected by GBP, even when we increased the total dosage and frequency
from once a day
to three times per day (see below and Experimental Procedures), and even
though an inbred
strain of mice was used. Nevertheless, these findings show that GBP is a
powerful inhibitor of
new synapse formation both in vitro and in vivo.
Example 6-Inhibition of TSP-induced synapse formation interferes with lesion-
induced barrel
cortex plasticity in neonatal mice
[02581 Understanding how the brain remodels its neuronal networks has been a
major goal
of neurobiology, as these processes underlie learning, memory and recovery
from injury. Is
astrocyte-induced synapse formation involved in remodeling neural circuits
during development?
To explore this question a well-established developmental plasticity paradigm,
`the barrel cortex
plasticity' assay, was used. The nerves that innervate the major whiskers on
the snout of the
mouse project to the brain as a topographically ordered "somatotopic" map
where afferent axons
and target cells form ordered modules that recapitulate the structural
organization seen on the
whisker pad (Erzurumlu et al., The Anatomical Record 288:121-134, 2006).
First, neurons of the
trigeminal nerve innervate the mystacial vibrissae (whiskers) and send
projections that crossover
completely to the opposite side of the brain. These projections then form
synapses at the

73


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
brainstem (barrelettes) and their afferents then project and synapse at the
thalamus (barreloids).
Finally, thalamocortical axons project to the primary somatosensory cortex,
where they form a
somatotopic map of "barrels" with postsynaptic layer IV granule cells (Figure
9A). To test
whether TSP-induced synapse formation is involved in mechanisms of experience-
dependent
plasticity, we took advantage of the ability of the barrel cortex to exhibit
structural changes of its
circuitry in response to peripheral whisker manipulations. If a row of
whiskers on the snout are
deafferentated during a critical period of postnatal development (the first 3
postnatal days in
mice), barrels in the cortex corresponding to the lesioned row of whiskers
shrink and fuse
together, while cells in neighboring barrels invade the territory vacated by
projections from the
lesioned whiskers (Van der Loos and Woolsey, Science 179:395-546, 1973). These
changes can
be visualized by analyzing the barrel cortex on postnatal day 7 (P7) (Figure
9A).
[02591 Two groups of neonatal mice were injected either with GBP or an equal
volume of
saline daily starting at P0 until P7. On P1, five whiskers from the C-row on
one side of each
mouse snout were surgically removed and cauterized. The mice were then
sacrificed at P7 and
their barrel structure corresponding to both the unlesioned "control"
hemisphere and the lesioned
hemisphere were analyzed. Both saline and GBP injected mice had typical
barrels formed on the
control side (Figure 9B top two left panels). On the lesioned side, however,
while all saline
injected mice displayed a typical barrel cortex plasticity pattern, where the
C row barrels
corresponding to the cauterized whiskers are fused and the neighboring B and D
row barrels
enlarge to innervate the vacated regions. 50% of the GBP injected mice
displayed an atypical
plasticity response (Figure 9B, right panels). In these mice, not only the C
row, but also the A
and B rows also lost form and fused, even though the analysis of the snouts
showed that the
whisker follicles for these rows were still present and undisturbed in these
mice (Figure 9B and
9C, for a collection of phenotypes please see Figure 10).
[0260] Because GBP strongly blocks TSP and astrocyte mediated synaptogenesis,
these
findings suggest that astrocyte secreted TSPs induce the synapse formation
required for rewiring
of the barrels post injury. Blocking TSP-induced synapse formation may lead to
the loss of a
"stop and connect" signal, thus the axons still continue to seek for an
appropriate target which
results in failure of the barrels to reform. In order to more directly test
the role of TSPs, barrel
cortex plasticity in TSP1/2 double knockout (KO) mice was examined. A third of
the TSP1/2K0
mice analyzed showed a very similar, aberrant barrel cortex plasticity
phenotype (Figure 9B,

74


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
bottom right panel), a pattern never observed in any of the wild type mice.
These findings
provide evidence that the main effect of GBP in barrel cortex plasticity is
likely through its
inhibition of astrocyte derived TSP-induced synapse formation and that TSP-
induced synapse
formation participates in barrel cortex plasticity in mice. Interestingly,
neither the GBP injected
mice nor the TSPI/2 KO mice had problems in the normal establishment of the
barrels in the
unlesioned-control side, suggesting that TSPs specifically play a role in
synaptic remodeling-
plasticity upon injury in this system.
Example 7-The calcium channel subunit aZSl interacts with the synaptogenic
domain of TSP
[0261] Many domains of TSP have previously been found to interact with
specific cell
surface receptors, particularly integrins, however until recently there were
no known receptors
for the EGF-like domains of TSPs. Recently, the EGF-like domains of TSP4 were
found to bind
to the VWF-A domain of integrin aM (Pluskota et al., Blood, 106:3970-3978,
2005). As several
other integrin a and (3 subunits contain VWF-A like domains (Whittaker and
Hynes, Mol. Bio.
of the Cell 13:3369-3387, 2002), we investigated whether integrin aM or the
other VWF-A
domain containing integrins were expressed by RGCs and were involved in TSP
induced
synapse formation. None of the integrins that contained the VWF-A domain and
were expressed
by RGCs were crucial for the synaptogenic activity of TSP (data not shown).
[0262] Another class of neuronal plasma membrane molecules that contains VWF-A
domains is the calcium channel subunit alpha2 delta (a28) family. Four a26
subunits in
mammals have been cloned to date (Klugbauer et al., J. of Bioenergetics and
Biomembranes 35:
639-647, 2003). Gene expression profiling of RGCs showed high level expression
of the L-type
calcium channel subunit a281, which was verified by RT-PCR and by Western
blotting (data not
shown, Figure l 1A). We therefore next investigated whether a281 interacts
with TSPs. We used
specific polyclonal antibodies to immunoprecipitate TSPs 1, 2 and 4 from
postnatal day 5 rat
cerebral cortex lysate and performed Western blot analysis on the
immunoprecipitated proteins
using a monoclonal antibody specific for a28 1. a281 was detected in
immunoprecipitations
performed using each of the three TSP antibodies (Figure 11B) providing
evidence for an
interaction between a281 and TSPs in vivo.
[0263] In order to determine whether there is a direct and specific binding
interaction
between the synaptogenic domain of TSP and a28 1, we co-expressed a FLAG-
tagged a281


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
alone (Figure 11 C lane 1), with SD2 (lane 2) or with an unrelated secreted
control protein that
was sub-cloned into the same vector as SD2 and had the same C-terminal myc and
6-Histidine
tags (Control-myc-his, Figure 11C lane 3). When we immunoprecipitated a251-
FLAG by using
beads that are conjugated to anti FLAG-tag antibody, we saw that SD2 co-
immunoprecipitated
with a281-FLAG but the Control-myc-his protein did not (Figure 11 C, lanes 5
and 6,

respectively). These data show that a251 specifically interacts with the
synaptogenic EGF-like
domains of TSP.
[0264] To provide further evidence for the interaction between a281 and SD2,
we co-
expressed a251 and SD2 in HEK293 cells and this time immunoprecipitated SD2
from detergent
solubilized HEK293 cell membrane preparations using beads conjugated with anti-
myc antibody.
a281 co-immunoprecipitated with SD2 further showing that a281 directly
interacts with the
synaptogenic domain of the TSPs (Figure 11D, lane 7). To determine whether the
other calcium
channel subunits were required for the SD2-a251 interaction, and whether the
calcium channel

a 1 subunit interacts with SD2, we co-expressed SD2 with the L-type calcium
channel a 1 C and 0
subunits in the presence or absence of a261(lanes 1,2 and 4). The a 1 subunit
did not interact
with SD2 even in the presence of a281 (Figure 11D, lanes 5,6 and 8), and the
amount of a281
that co-immunoprecipitated with SD2 decreased when the al subunit was also
expressed (Figure
3D, lane 8 top, panel). Thus the synaptogenic domain of TSP specifically
interacts with a281 but
not with the al subunit of the L-type calcium channel. The a2S1-SD2
interaction was strongly
dependent on the presence of magnesium ions in the buffers used for membrane
preparation and
immunoprecipitation (Experimental Procedures). In conclusion, these data show
that a261 and
TSP interact through the synaptogenic domain of TSP and this interaction does
not depend on
the presence of other L-type calcium channel subunits.
Example 8-Preparation and testing of the TSP2 synaptogenic domain construct
SD2
[0265] A mammalian expression construct designed to encode a secreted
monomeric TSP2
fragment that encompassed the third properdin-like repeat and the three EGF-
like repeats of
TSP2 was made (Figure 12A). This recombinant protein from transfected HEK293
cell culture
media to homogeneity by utilization of the C-terminal 6-Histidine tag (His-
tag) was purified
(Figure 12B). This tagged and purified TSP2 fragment (designated SD2 for
synaptogenic domain
2) was strongly synaptogenic (Figure 12C, D). These synapses mimicked the
synapses induced

76


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
by full length TSP1, as they were presynaptically active, but postsynaptically
silent (data not
shown). The activity of SD2 was highest in the 10-20 nM range (Figure 12D). At
higher
concentrations its activity was diminished suggesting the inhibition or
desensitization of its
receptor due to high ligand concentration.
Experimental Procedures for Examples 2-8
Purification and Culture of RGCs and astrocytes
[0266] RGCs were purified by sequential immunopanning to greater than 99.5%
purity
from P5 Sprague-Dawley rats (Charles Rivers) and cultured in serum-free medium
containing
BDNF, CNTF, and forskolin on laminin-coated coverslips, as previously
described
(Christopherson et al., Cell 120:421-433, 2005; Meyer-Franke et al., Neuron
15:805-819, 1995;
Ullian et al., Science 291:657-661, 2001). Cortical astrocyte inserts and ACM
were prepared as
described in (Christopherson et al., Cell 120:421-433, 2005). RCGs were
cultured for 3-4 days
to allow robust process outgrowth and then cultured with astrocyte inserts,
ACM or TSPs for an
additional 6 days.
Recombinant proteins and DNA constructs
[0267] Purified human platelet TSPI was obtained from Haematologic
Technologies.
Recombinant TSP4 and TSP5 were expressed and purified as described in (Chen et
al., J. Biol.
Chem. 275:26538-26544, 2000; Lawler et al., J. Biol. Chem. 270:2809-2814,
1995). Mouse
TSP3 cDNA in pcDNA3 mammalian expression vector was a gift from V. Dixit
(Qabar et al., J.
Biol. Chem. 269:1262-1269, 1994). TSP3 was overexpressed in Cos7 cells and the
Cos7 cell
conditioned media was used as a source of TSP3. In the same experiment the
control conditions
were treated with conditioned media from Cos7 cells transfected with empty
pcDNA3 vector.
The panel of TSP 1 and 2 truncation constructs were expressed and purified as
described before
in (Mosher et al., Methods in Cell Bio. 69;69-81, 2002; Miao et al., Cancer
Research 61:7830-
7839, 2001; Saumet et al., Blood 106:658-667, 2005).
[0268] Overexpression vector for a281 was a kind gift from D. Lipscombe (Brown
University). 81 expression vector was a kind gift from K. Campbell (Univ. of
Iowa) and is
described in (Gurnett et al., J. Biol. Chem. 272:18508-18512, 1997). Vectors
expressing calcium
channel subunits alC and 0 are described in (Dolmetsch et al., Science 294:333-
339, 2001).
[0269] The synaptogenic domain of TSP2 (SD2) was cloned into pAPtag5 vector
(GenHunter) between SfiI and XhoI sites. SD2 was expressed by HEK293 cells,
which were

77


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
transfected using Lipofectamine 2000 (Invitrogen) following manufacturers
instructions. The
secreted recombinant protein was then purified from conditioned culture media
by Ni-chelating
chromatography using Ni-NTA resin (Qiagen) following manufacturers
instructions.
Synapse Assay on RGCs
[0270] For synapse quantification of RGC cultures cells were fixed for 7
minutes with 4%
paraformaldehyde (PFA), washed three times in phosphate-buffered saline (PBS),
and blocked in
100 l of a blocking buffer containing 50% normal goat serum and 0.1% Triton X-
100 for 30
minutes. After blocking, coverslips were washed three times in PBS, and 100 L
of primary
antibody solution was added to each coverslip, consisting of rabbit anti-
synaptotagmin (1:750,
cytosolic domain, Synaptic Systems) and mouse anti-PSD-95 (1:750, 6G6-1C9
clone, Affinity
Bio Reagents). Coverslips were incubated overnight at 4 C, washed three times
in PBS, and
incubated with 100 l of Alexa-594 conjugated goat anti-rabbit and Alexa-488
conjugated goat
anti-mouse (Invitrogen) diluted 1:1000 in antibody buffer. Following
incubation for 2 hours,
coverslips were washed 3-4 times in PBS and mounted in Vectashield mounting
medium with
DAPI (Vector Laboratories Inc) on glass slides (VWR Scientific). Secondary-
only controls were
routinely performed and revealed no significant background staining.
[0271] Mounted coverslips were imaged using Nikon Eclipse E800 epifluorescence
microscope (Nikon). Healthy cells that were at least two cell diameters from
their nearest
neighbor were identified and selected at random by eye using DAPI
fluorescence. Eight bit
digital images of the fluorescence emission at both 594 nm and 488 nm were
recorded for each
selected cell using a monochrome CCD camera and SPOT image capture software
(Diagnostic
Instruments, Inc). Merged images were analyzed for co-localized puncta by
using a custom-
written plug-in (Barry Wark, licensed under the GPL (see worldwide web at
gnu.org/copyleft/gpl.html) for the NIH image processing package ImageJ (see
worldwide web at
rsb.info.nih.gov.laneproxy. stanford.edu/ij/). This analysis generates counts
that are similar to the
numbers obtained counting by eye was verified numerous times. It has been
previously shown
that increase in co-localized puncta corresponds to a real increase in the
number of synapses,
which were counted by electron microscopy and confirmed by
electrophysiological analysis
(Christopherson et al., Cell 120:421-433, 2005; Ullian et al., Science 291:657-
661, 2001).

78


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
RGC Transfections
[0272] 6DIV RGCs were transfected using the Lipofectamine 2000 (Invitrogen)
reagent.
Briefly, 300 l conditioned culture medium from the cells was removed and
saved in another
tissue culture plate at 37 C in 10% C02 incubator. The cells were then fed
with 200 l fresh
media. 1 g DNA or 2 l of 20 M siRNA pool was mixed with 100 l OptiMEM
media
(Invitrogen) and 2 l Lipofectamine 2000 reagent (Invitogen). The mixture was
incubated for 20
minutes at RT and was added to the cells. After 3 hours cells were washed
twice with warm PBS
and were fed with 200 l of fresh RGC growth media and 300 l of the saved
conditioned
media. SD2 or astrocyte insert treatments started 1 day post-transfection for
plasmid constructs
and 2 days post-transfection for siRNA pools. Transfected cells were marked by
GFP that was
co-transfected with each condition. Typical transfection efficiencies ranged
between 10% and
15%. Cells were stained for synapses after 6 days of SD2 or astrocyte
treatment as described
before. In this case PSD95 was detected by using a secondary goat anti-mouse
antibody
conjugated to Alexa 680. Images of transfected cells were taken in three
channels (488nm for
GFP, 594nm for synaptotagmin, and 680 nm for PSD95). The number of synapses on
GFP
positive cells was quantified using methods described above.

Immunoprecipitations and Western Blotting
[0273] SD2, a251(untagged or FLAG-tagged) and L-type calcium channel a I C and
(3
subunits were expressed in HEK293 cells by transient transfection using
Lipofectamine 2000
(Invitrogen) following the provider's instructions, for 2 to 3 days.
[0274] HEK293 cell plasma membranes were prepared for co-immunoprecipitations
as
follows. Cells were washed with PBS 3 times and scraped from the tissue
culture plates. The
cells were pelleted and resuspended in ice-cold hypotonic buffer (10mM Tris pH
7.4, 1 mM
CaC12 and 1mM MgC12) with protease inhibitors (Complete EDTA-free, Roche), and
incubated
on ice for 15 minutes for cells to swell. The cells were then disrupted by
homogenization in a
glass-on-glass douncer (5 times). The nuclei and unbroken cells were removed
by centrifugation
at 300g for 5 minutes. The post-nuclear supernatant was centrifuged for 20
minutes at 20,000g to
pellet the membranes.
[0275] The membranes were resuspended in solubilization buffer (25mM Tris pH
7.2,
150mM NaCI, 250mM Sucrose, 1mM CaCl2 and ImM MgC12) with protease inhibitors

79


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
(Complete EDTA-free, Roche) and 0.5% Surfact-Amps NP-40 (Pierce) and were
incubated at
4 C for 10 minutes to allow for solubilization. The insoluble debris was
removed by
centrifugation (20,000g for 10 minutes). The supernatant was incubated with
anti-myc antibody
conjugated agarose beads to immunoprecipitate SD2 (Upstate) or with anti-FLAG
antibody M2
conjugated beads to immunoprecipitate FLAG-tagged a281(Sigma-Aldrich) for 4
hours or
overnight at 4 C while rotating. After binding was completed the beads were
washed 4-5 times
with the solubilization buffer. The bound proteins were eluted by addition of
non-reducing SDS-
PAGE buffer (2X, Pierce) and 5 min incubation at 37 C. The eluate was then
transferred to a
clean tube and (3-mercapto-ethanol was added. The samples were denatured at 37
C for 30
minutes and loaded on SDS-PAGE gels (4-15%, BioRad). After SDS-PAGE
electrophoresis,
proteins were transferred onto PVDF membranes and were blotted for target
proteins.
[0276] TSP immunoprecipitations were carried out as follows. Five P5 rat
cortices were
dissected and membranes were prepared and solubilized as described above. The
soluble fraction
was incubated with protein A/G beads that were pre-bound to TSP 1, 2 or 4
polyclonal antibodies
overnight (the TSP antibodies are described in (Lawler et al., J. Biol. Chem.
270:2809-2814,
1995; Tooney et al., Matrix Biol. 17:131-143, 1998)). The beads were washed 4-
5 times and the
proteins were eluted and prepared for SDS-PAGE analysis as described above.
[0277] The calcium channel subunit a281 was detected in Western blots by using
a
monoclonal antibody (Sigma, 1:1000). SD2 and the myc-tagged control protein
both contained C
terminal 6-His tags and they were recognized in Western blot by using a
monoclonal anti-penta-
Histidine antibody (Qiagen, 1:1000). Calcium channel subunit alC was detected
with a rabbit
polyclonal antibody (Chemicon, 1:1000). Horseradish peroxidase conjugated anti-
mouse or anti-
rabbit (1:5000) were used as secondary antibodies (Jackson Labs) and the
detection was
performed with an ECL kit from Amersham.

Synapse assay on mouse brain sections
[0278] Brains were immersed in 4% paraformaldehyde (PFA), fixed overnight at 4
C, and
cryo-protected in 30% sucrose. For synaptic staining, tissue was embedded in a
2:1 mixture of
20% sucrose:OCT in PBS, and cryo-sectioned (12 m). Sections were dried at 37
C, washed
three times in PBS, and blocked with 20% normal goat serum (NGS, Invitrogen)
in PBS 1 hr.
Primary antibodies were diluted in PBS with 0.3% triton and 10% NGS as
follows: PSD95



CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
(Zymed, rabbit, 1:500) and VGlutl and VGlut2 (Chemicon, guinea pig, 1:2500)
incubated
overnight at 4 C. Secondary Alexa-conjugated antibodies (goat anti-guinea pig
Alexa 488 and
goat anti-rabbit Alexa 594, Invitrogen) were added at (1:200 in same buffer)
for 2 hr at room
temperature in dark. Slides were mounted in Vectashield with DAPI and were
imaged using a
Zeiss LSM 510 confocal laser-scanning microscope.
[0279] Three independent sagital brain sections per animal were stained with
pre- and post-
synaptic markers and 5 m confocal scans were performed (optical section width
0.38 m, 14
optical sections each) at the cortex. To ensure consistency of the cortical
area to be scanned, the
outer cortical region, including the synaptic layer of the cortex dorsal to
the dentate-gyrus were
picked in each section. The parameters for scanning were always setup for wild-
type (or saline
injected) brain sections and same imaging parameters were used for transgenic
(or GBP injected)
animals. Merged single optical section images at 1 m intervals were analyzed
using the ImageJ-
puncta analyzer option to count for number of co-localized pre- and post-
synaptic puncta (5
optical sections per brain section and 15 total images per brain). Average
synaptic density per
imaged area was calculated for each condition.

Mice
[0280] TSP1/2 double knockout mice on an FVB background were used (n=12) (Agah
et
al., Matrix Biol. 22:539-47, 2004). Wildtype mice with FVB background were
purchased from
Charles River Laboratories. Brains from P21, a281 overexpressing, transgenic
animals and their
littermate wild-type controls (n=8) were provided by Li and colleagues and are
described in (Li
et al., Pain 125:20-34, 2006).

Saline and gabapentin injections:
[0281] In three independent experiments, two litters of wild type mice
(n=3x12, FVB
background) were given daily intraperitoneal injections of either a single
dose of 400mg/kg of
GBP (Sigma-Aldrich) or a matching volume of saline solution (PBS). In one
experiment the
mice were injected with 200mg/kg GBP (n=6) three times a day and the controls
received the
same volume of saline solution (n=6). Pups were weighed just before injections
to determine the
dose administered and also to follow their weight gain and general health,
which showed no

81


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
differences between GBP and saline injected mice. Samples were blinded during
analysis of the
barrel cortex plasticity.

Whisker lesions:
[0282] All whisker lesions removed whisker follicles from the center row (row
C) on the
right whisker pad of P 1 mice. Neonatal mice were held on their left side
under a dissecting scope
and received two parallel incisions with a surgical blade flanking the row of
whiskers to be
removed. The skin between the incisions was pulled back with forceps.
Follicles were
individually removed with forceps at the opening. The lesion site was then
cauterized with silver
nitrate using flexible caustic applicators (Tech-Med). Mice were then allowed
to recover in their
home cage.

Barrel cortex immunohistochemistry:
[0283] Mice were sacrificed at P7 and brains were dissected and immersion
fixed in 4%
paraformaldehyde for 12 to 24 hours. Brains were cryoprotected in 30% sucrose
in PBS until
they sank to the bottom of the solution (24-48 hours). Brains were then
hemisected along the
midline, and 40- m-thick sections were cut tangential to the barrel cortex of
each hemisphere on
a Leica SM2000R freezing microtome and placed in PBS. Barrel cortex staining
was performed
on free-floating sections. First, sections were placed in a blocking solution
of 10% NGS and
0.25% Triton-X-100 in PBS for 45 minutes at RT. Sections were then incubated
with anti-
serotonin (5-HT) transporter rabbit polyclonal antibody (Calbiochem) at 1:400
in blocking
solution for one hour at room temperature, and then overnight at 4 C. After 3
washes in PBS,
tissue sections were incubated with goat anti-rabbit Alexa 594 secondary
antibody (Invitrogen) at
1:1000 in blocking solution for 90 minutes at room temperature to allow
fluorescent detection of
the primary antibody. After 3 washes in PBS, sections were mounted on glass
slides (VWR), in
the order in which they were cut, and mounted with Vectashield Mounting Medium
(Vector
Laboratories). Sections were visualized using a Nikon Eclipse E800 fluorescent
microscope, and
images were digitally acquired using an SPOT camera (Diagnostic Instruments,
Sterling Heights,
MI).

82


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
Barrel cortex reconstructions:
[0284] To reassemble complete maps of the barrel cortex, 5-HTT stained images
from serial
sections were reconstructed in Photoshop (Adobe Systems). One section, usually
with most of
the barrel cortex already in plane, was selected as the base image upon which
data from adjacent
sections were layered. Each added section was first carefully aligned to match
the previous
sections, and then only the portions of the barrel field labeled more brightly
in added sections
were revealed with a layer mask. This process was repeated until the entire
primary barrel cortex
was visible. Contrast and brightness levels were then carefully matched
between layers to attain
a more accurate representation of the level of signal across the entire
reconstructed barrel field.
Whisker pad staining:
[0285] The whole snout region was immersion fixed in 4% paraformaldehyde and
then
cryoprotected in 30% sucrose in PBS overnight. Snouts were hemisected along
the midline, and
trimmed back to only contain the whisker pads. Whisker pads were then
flattened and embedded
in 2:1 30% sucrose:O.C.T. Sections were cut at 501im and collected on positive
coat slides
(Sigma) using a Leica CM3050 cryostat. Sections were dried at 37 C for 30 to
90 minutes, and
then washed with PBS once and stained with Mayer's Hematoxylin (Lillie's
Modification)
(Dako) for 30 seconds. Slides were rinsed in deionized distilled water and
coverslipped with
Faramount aqueous mounting medium (Dako). Brightfield microscopy was performed
with the
Nikon Eclipse E800 microscope, and images were digitally acquired using an
SPOT camera
(Diagnostic Instruments).
Example 9- Intrathecal bolus injection of active anti-TSP4 antibody or TSP4
antisense
oligodeoxynucleotides reverses allodynia in spinal nerve injured rats
[0286] To test if TSP4 protein could induce behavioral hypersensitivity and
gabapentin
could block the induction, the following experiments were carried out. Active
or heat inactivited
TSP4 protein dissolved in saline was injected on day 0, and behavioral
hypersensitivity test was
conducted daily as described in the Experimental Procedures. Gabapentin or
saline was injected
three days after TSP4 (45 ug/rat) bolus injection, followed by behavioral test
1 hour later and
then daily thereafter. As shown in Figure 13A, intrathecal bolus injection of
active, but not heat-
inactivated TSP4 protein into L5/6 spinal cord segments of naive rats caused a
gradual reduction
in their hindpaw withdrawal thresholds to von Frey filament (mechanical)
stimulation in a dose-

83


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
dependent manner (Figure 13A). The behavioral hypersensitivity had an onset
time of two days
post injection and peaked about 2-4 days post injection, and lasted over a
week post TSP4. TSP4
induced behavioral hypersensitivity was reversible, and the reversal time was
about five days
post the peak effect time. The behavioral hypersensitivity induced by TSP4 was
blocked by
intrathecal injection of gabapentin (1 mg/rat, bolus i.t. injection), but not
saline. The gabapentin
effects lasted over one day. Since gabapentin binds to the calcium channel
a281 subunit, these
data support that TSP4-induced allodynia is likely mediated by interacting
with the calcium
channel a281 subunit. The slow reversal of the gabapentin effects suggested a
chronic, rather
than acute, mechanism of the drug actions.
[0287] To determine if TSP4 plays a role in mediating neuropathic pain
processing at the
spinal cord level, the effect of intrathecal TSP4 antisera in reversing
established allodynia in
spinal nerve ligated rats was examined. Intrathecal bolus injection of active
TSP4 antibody
(chicken polyclonal, from Dr. Frank Zaucke at University of Cologne, Cologne,
Germany) was
administered into left L5/6 spinal nerve ligated rats 2-weeks post injury,
when the injured rats
had fully developed allodynia. The bolus TSP4 antibody reversed established
allodynia at the
injury side in a dose-dependent manner (Fig. 13B-C). Injection with the heat-
inactivated (boiled)
anti-TSP4 antibody did not show any effect in allodynia reversal (Fig. 13B-C).
The effects of
active antisera (80 ug/rat, bolus i.t. injection) in allodynia reversal had an
onset time of 4 hrs and
duration of over 10 hrs (Fig. 13B). Similar active TSP4 antibody treatment did
not change the
base line behavioral thresholds to the same stimuli in the non-injury
(contralateral) side. These
data suggest that elevated TSP4 protein may play a critical role in peripheral
nerve injury-
induced allodynia at the spinal level, and blocking its functions at the
spinal level may have
therapeutic values in pain management.
[0288] To determine if similar anti-TSP4 treatment could prevent the
development of
injury-induced allodynia, spinal nerve ligated rats were treated pre-emptively
with intrathecal
daily injection of TSP4 antisera (chicken polyclonal, 80 ug/rat). The
treatment started before the
nerve ligation surgery. As shown in Fig. 13D, pre-emptive intrathecal TSP4
antiserum treatment
did not alter the base line thresholds in the non-injury (right) side, but
prevented the onset of
allodynia in the injury (left) side of spinal nerve ligated rats in comparison
to the injured rats
treated with intrathecal injection of saline. The TSP4 antibody effects lasted
over 2 days after
the last injection. These data that injury-induced TSP4 protein may play a
critical role in the

84


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
initiation of allodynia at the spinal level and blocking TSP4 function may
have therapeutic
benefits in neuropathic pain prevention.
[0289] To determine if TSP4 antisense oligodeoxynucleotides can block injury-
induced
tactile allodynia, two TSP4 antisense oligodeoxynucleotides (#1 and #2)
complementary to two
different regions of the TSP4 mRNA were intrathecally injected into spinal
nerve ligated rats 5
weeks post injury when the injured rats had established allodynia at the
injury side. As shown in
Fig. 13E, daily intrathecal injection of TSP4 antisense #1 (50 ug/day) for
four days had some
effects in allodynia reversal, but similar treatment with antisense, not
mismatch #2, caused a
complete reversal of established allodynia in the injury (ipsilateral) side to
a level similar to that
observed in the non-injury (contralateral) side. The antisense effects had a 3-
day onset time,
peaked one day after the 4-day treatment, and lasted for 3 days after the last
injection. Neither
the antisense nor mismatch oligodeoxynucleotides altered the behavioral
thresholds at the non-
injury side. These data that blocking injury-induced TSP4 expression with
antisense
oligodeoxynucleotides may represent another approach in blocking the effects
of spinal TSP4 in
pain processing.

Experimental Procedures for Example 9
Surgery and behavioral testing
[0290] Male Harlan Sprague Dawley rats (100-150 g, Harlan Industries
Indianapolis, IN)
were housed in separate cages, exposed to a 12/12 h day/night cycle and
allowed free access to
food and water. Peripheral nerve injury was induced in isoflurane-anesthetized
animals with
tight ligation of the left L5/6 spinal nerves between DRG and the beginning of
the spinal nerve
(Kim and Chung, Pain 50 50:355-363, 1992). To access tactile allodynia, paw
withdrawal
threshold (PWT) was determined using the up-down method of Dixon (Dixon, Ann.
Rev.
Pharma. & Toxico. 20:441-462, 1980) with von Frey filaments (Stoelting, Wood
Dale, IL, USA)
as described previously (Chaplan et al., J. Neuros. Methods 53:55-63, 1994).
Briefly, animals
were allowed to acclimate for 30 min in a clear plastic cubicle with a wire-
mesh base. A 2 g
calibrated filament was first applied to the left hindpaw plantar surface with
a pressure causing
the filament to bend. If no paw lifting was detected after 5s the next
filament with increasing
weight was used. If a paw lifting was observed, the next weaker filament was
used. This
paradigm was used for a total of six measurements, starting from the one
before the first change



CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
in response, or five responses (assigned a score of 0.25 g) were reached. The
50% response
thresholds were calculated as described previously (Luo et al., J. Neurosci.
21:1868-75, 2001).
Intrathecal antisense oligodeoxynucleotides
[0291] An antisense oligodeoxynucleotide #1 against the translation initiation
region of the
target gene was designed and has been shown to be effective in knocking down
the expression of
other target genes (Wahlestedt et al., Nature 363:260-263, 1993; Ji et al.,
PNAS 91:12540-
12543, 1994; Hua et al., J. Neurochem. 70:688-698, 1998; Li et al., J.
Neurosci. 24:8494-8499,
2004). Oligodeoxynucleotide #2 based on a segment of rat TSP4 mRNA sequence
was also
designed and has been used for producing primers in real-time PCR experiments
(TaqMan Gene
Expression Assay ID: Rn014934317, Applied Biosystems), and is specific to the
TSP 4 mRNA.
Mismatch oligodeoxynucleotides, which contain the same number of nucleotides
but in a random
order (not complementary to any particular region of TSP4 mRNA), were used as
controls.
Since a single base mismatch would reduce binding affinity about 500-fold
(Freier, 1992), the
mismatch oligodeoxynucleotides should have very low affinity to TSP4 mRNA. The
nucleic
acid sequences for these regions of rat TSP4 gene, and the antisense and
mismatch
oligodeoxynucleotides are shown below:

Rat TSP4 cDNA sequence corresponding to the translation initiation region
(initiation codon
underlined):
5' CgtatgAccATGAttAcgCC 3' (SEQ ID NO: 1; Genbank accession #: X89963)
Oligodeoxynucleotides:
Antisense #1: 5' GGCGTAATCATGGTCATACG 3' (SEQ ID NO:2)
Mismatch #1: 5' CGGAGTCATGATCGTAATCG 3' (SEQ ID NO:3)

Rat TSP4 cDNA sequence corresponding to the region for the real-time PCR
primers:
5'GGAAGATAGCAACAATGATGG 3' (SEQ ID NO:4; Genbank accession #: X89963)
Oligodeoxynucleotides:

Antisense #2: 5' CCATCATTGTTGCTATCTTCC 3' (SEQ ID NO:5)
Mismatch #2: 5' ACCATCGTTGTTACTTTCTCC 3' (SEQ ID NO:6)
86


CA 02697363 2010-02-22
WO 2009/029173 PCT/US2008/009747
[0292] These antisense and mismatch sequences were not complementary to any
sequences
of other rat genes as indicated by BLAST search results in rat genome
database. It had been
shown that oligodeoxynucleotides with phosphorothioate modification have
increased potential
for crossing lipid bilayers, and reduce nuclease cleavage (Crooke, et al.,
Ann. Rev.
Pharmacology. & Toxicology, 1996). Limited phosphorothioate modification on
only three
nucleotides at both ends of the oligodeoxynucleotides was used, and this
method of modification
does not cause inflammatory side effects in vivo (Li et al., J. Neurosci.
24:8494-8499, 2004). All
antisense oligodeoxynucleotides used in the studies were synthesized
commercially (Genelink,
Inc., NY), sterilized with ethanol precipitation, and dissolved in sterile
saline before use.
Drug preparation and intrathecal injection
[0293] Gabapentin, purified TSP proteins and antibodies are dissolved and
diluted in sterile
saline before use and directly injected into the L5/6 spinal region of an
isofluorane anesthetized
rat in a total volume of 10 mL using a microinjector connected to a 30 G
needle.
Statistics
[0294] Unpaired Student's t tests were performed, and significance was
indicated by a two-
tailed p value of < 0.05.
[0295] All publications and patent applications cited in this specification
are herein
incorporated by reference as if each individual publication or patent
application were specifically
and individually indicated to be incorporated by reference. The citation of
any publication is for
its disclosure prior to the filing date and should not be construed as an
admission that the present
invention is not entitled to antedate such publication by virtue of prior
invention.
[0296] It is to be understood that this invention is not limited to the
particular methodology,
protocols, cell lines, animal species or genera, and reagents described, as
such may vary. It is
also to be understood that the terminology used herein is for the purpose of
describing particular
embodiments only, and is not intended to limit the scope of the present
invention, which will be
limited only by the appended claims.
[0297] All technical and scientific terms used herein have the same meaning as
commonly
understood to one of ordinary skill in the art to which this invention belongs
unless clearly
indicated otherwise.

87

Representative Drawing

Sorry, the representative drawing for patent document number 2697363 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2008-08-14
(87) PCT Publication Date 2009-03-05
(85) National Entry 2010-02-22
Dead Application 2013-08-14

Abandonment History

Abandonment Date Reason Reinstatement Date
2012-08-14 FAILURE TO PAY APPLICATION MAINTENANCE FEE

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2010-02-22
Maintenance Fee - Application - New Act 2 2010-08-16 $100.00 2010-07-16
Registration of a document - section 124 $100.00 2011-02-16
Maintenance Fee - Application - New Act 3 2011-08-15 $100.00 2011-07-14
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
THE BOARD OF TRUSTEES OF THE LELAND STANFORD JUNIOR UNIVERSITY
Past Owners on Record
BARRES, BEN A.
EROGLU, CAGLA
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 2010-02-22 87 5,071
Drawings 2010-02-22 14 218
Claims 2010-02-22 6 196
Abstract 2010-02-22 1 53
Cover Page 2010-05-12 1 29
Description 2010-05-14 87 5,071
Correspondence 2010-05-25 2 58
PCT 2010-02-22 2 99
Assignment 2010-02-22 3 79
Correspondence 2010-05-08 1 19
Prosecution-Amendment 2010-05-14 2 78
Assignment 2011-02-16 5 250
Correspondence 2011-12-21 3 83
Assignment 2010-02-22 5 129

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :